You are on page 1of 413

5IF"VTUSBMJBO



S A T

UI&EJUJPO




M M U N

)BOECPPL

*NNVOJTBUJPO

O N

RECOGNITION AND TREATMENT OF ANAPHYLAXIS


Signs of anaphylaxis
Anaphylaxis causes respiratory and/or cardiovascular signs or symptoms AND involves other organ systems such as skin or GI tract, with:
t TLJOTJHOT TVDIBTUIFSBQJEEFWFMPQNFOUPGVSUJDBSJBMMFTJPOTPSFSZUIFNB
t TJHOTPGVQQFSBJSXBZPCTUSVDUJPO TVDIBTIPBSTFOFTTBOETUSJEPS
t JOEJDBUJPOTPGMPXFSBJSXBZPCTUSVDUJPO TVDIBTTVCKFDUJWFGFFMJOHTPGSFUSPTUFSOBMUJHIUOFTT EZTQOPFBPSXIFF[F
t MJNQOFTTBOEQBMMPS XIJDIBSFTJHOTPGTFWFSFBOBQIZMBYJTJODIJMESFO
t QSPGPVOEIZQPUFOTJPOJOBTTPDJBUJPOXJUIUBDIZDBSEJB BOEPSPUIFSTJHOTPGDBSEJPWBTDVMBS
EJTUVSCBODF TVDIBTTJOVTUBDIZDBSEJBPSTFWFSFCSBEZDBSEJB
t BCEPNJOBMDSBNQT EJBSSIPFBBOEPSWPNJUJOH
Management of anaphylaxis
t *GUIFQBUJFOUJTVODPOTDJPVT QMBDFIJNIFSPOUIFMFGUTJEFBOEQPTJUJPOUPLFFQUIFBJSXBZDMFBS*GUIFQBUJFOUJT
DPOTDJPVT QMBDFTVQJOFJOAIFBEEPXOBOEGFFUVQQPTJUJPO VOMFTTUIJTSFTVMUTJOCSFBUIJOHEJDVMUJFT

t (JWFBESFOBMJOFCZJOUSBNVTDVMBSJOKFDUJPO TFFCFMPXGPSEPTBHF
GPSBOZTJHOTPGBOBQIZMBYJTXJUISFTQJSBUPSZBOE
PSDBSEJPWBTDVMBSTZNQUPNTPSTJHOT"MUIPVHIBESFOBMJOFJTOPUSFRVJSFEGPSHFOFSBMJTFEOPOBOBQIZMBDUJDSFBDUJPOT
TVDIBTTLJOSBTIXJUIPVUPUIFSTJHOTPSTZNQUPNT
BENJOJTUSBUJPOPGJOUSBNVTDVMBSBESFOBMJOFJTTBGF
t *GUIFSFJTOPJNQSPWFNFOUJOUIFQBUJFOUTDPOEJUJPOCZNJOVUFT SFQFBUEPTFT
PGBESFOBMJOFFWFSZNJOVUFTVOUJMJNQSPWFNFOUPDDVST
t *GPYZHFOJTBWBJMBCMF BENJOJTUFSCZGBDFNBTLBUBIJHIPXSBUF
t $BMMGPSQSPGFTTJPOBMBTTJTUBODFBOEDBMMBOBNCVMBODF/FWFSMFBWFUIFQBUJFOUBMPOF
t #FHJOFYQJSFEBJSSFTVTDJUBUJPOGPSBQOPFB DIFDLGPSBDFOUSBMQVMTF*GDFOUSBMQVMTF
OPUQBMQBCMF DPNNFODFFYUFSOBMDBSEJBDNBTTBHF &$.

t "MMDBTFTTIPVMECFBENJUUFEUPIPTQJUBMGPSGVSUIFSPCTFSWBUJPOBOEUSFBUNFOU
&YQFSJFODFEQSBDUJUJPOFSTNBZDIPPTFUPVTFBOPSBMBJSXBZJGUIFBQQSPQSJBUFTJ[FJTBWBJMBCMF CVUJUTVTFJTOPUSPVUJOFMZSFDPNNFOEFEVOMFTT
UIFQBUJFOUJTVODPOTDJPVT
"OUJIJTUBNJOFTBOEPSIZESPDPSUJTPOFBSFOPUSFDPNNFOEFEGPSUIFFNFSHFODZNBOBHFNFOUPGBOBQIZMBYJT
Adrenaline Dosage
5IFSFDPNNFOEFEEPTFPGBESFOBMJOFJTN-LHCPEZXFJHIU FRVJWBMFOUUP
NHLHVQUPBNBYJNVNPGN-PSNH
HJWFOCZEFFQJOUSBNVTDVMBSJOKFDUJPOJOUPUIFUIJHI
notUIFEFMUPJESFHJPO
"ESFOBMJOFmust not CFBENJOJTUFSFEJOUSBWFOPVTMZ
"ESFOBMJOFDPOUBJOTNHPGBESFOBMJOFQFSN-PGTPMVUJPOJOBN-HMBTTWJBM
5IFVTFPGBESFOBMJOFJTSFDPNNFOEFECFDBVTFJUJTVOJWFSTBMMZBWBJMBCMF6TFBN-
TZSJOHFUPJNQSPWFUIFBDDVSBDZPGNFBTVSFNFOUXIFOESBXJOHVQTNBMMEPTFT
5IFGPMMPXJOHUBCMFMJTUTUIFEPTFTPGBESFOBMJOFUPCFVTFEJGUIFFYBDUXFJHIUPGUIFJOEJWJEVBMJTOPULOPXO
%PTFTPG POFJOPOFUIPVTBOE
BESFOBMJOF
-FTTUIBOZFBS

oN-

oZFBST BQQSPYLH

N-

oZFBST BQQSPYLH

N-

oZFBST BQQSPYLH

N-

oZFBST BQQSPYLH

N-

ZFBSTBOEPWFS PWFSLH

N-

oZFBST BQQSPYLH

N-

'PSNPSFEFUBJMFEJOGPSNBUJPO TFF4FDUJPO Adverse events following immunisation

CONTACT DETAILS FOR AUSTRALIAN, STATE AND


TERRITORY GOVERNMENT HEALTH AUTHORITIES
Australian Government health authorities
Australian Government Health




State and Territory Government health authorities


"VTUSBMJBO$BQJUBM5FSSJUPSZ
*NNVOJTBUJPO&ORVJSZ-JOF




/FX4PVUI8BMFT

$POUBDUZPVSMPDBM1VCMJD)FBMUI6OJU GPVOEVOEFSA)FBMUIJOUIF8IJUF1BHFT

Northern Territory




Queensland




4PVUI"VTUSBMJB*NNVOJTBUJPO
$PPSEJOBUJPO6OJU



IUUQXXXEITBHPWBVQFIT

Tasmania


 or 
IUUQXXXEIITUBTHPWBVQVCMJDIFBMUIJNNVOJTBUJPOJOEFYIUNM

Victoria


IUUQXXXIFBMUIWJDHPWBVJNNVOJTBUJPO

8FTUFSO"VTUSBMJB




'PSJOGPSNBUJPOPOUIF"VTUSBMJBO$IJMEIPPE*NNVOJTBUJPO3FHJTUFSPSUPSFRVFTU
BDIJMETJNNVOJTBUJPOIJTUPSZ QMFBTFSFGFSUP"QQFOEJY

Whats new?
Changes introduced
in this edition of
the Handbook
See Chapter 1.1

COMPARISON OF THE EFFECTS OF DISEASES AND THE SIDE EFFECTS OF VACCINES


DISEASE

EFFECT OF DISEASE

SIDE EFFECT OF VACCINE

%JQIUIFSJBoDPOUBHJPVTCBDUFSJBTQSFBECZESPQMFUT
DBVTFTTFWFSFUISPBUBOECSFBUIJOHEJDVMUJFT

"CPVUJOQBUJFOUTEJFT5IFCBDUFSJBSFMFBTFBUPYJO 
XIJDIDBOQSPEVDFOFSWFQBSBMZTJTBOEIFBSUGBJMVSF

%51BE5QBWBDDJOFoBCPVUJOIBTMPDBMJOBNNBUJPOPS
GFWFS#PPTUFSEPTFTPG%51BNBZPDDBTJPOBMMZCFBTTPDJBUFEXJUI
FYUFOTJWFDJSDVNGFSFOUJBMTXFMMJOHPGUIFMJNC CVUUIJTSFTPMWFT
DPNQMFUFMZXJUIJOBGFXEBZT4FSJPVTBEWFSTFFWFOUTBSFWFSZSBSF

)FQBUJUJT"oDPOUBHJPVTWJSVTTQSFBECZDPOUBDUPSJOHFTUJPO
PGGBFDBMMZDPOUBNJOBUFEXBUFSGPPEPSUISPVHIDPOUBDUXJUI
UIFGBFDBMNBUFSJBMPGBQFSTPOJOGFDUFEXJUIIFQBUJUJT"

+BVOEJDF ZFMMPXJOHPGUIFTLJOBOEFZFT
GFWFS BOPSFYJB OBVTFB  "CPVUJOXJMMIBWFEJTDPNGPSUPSMPDBM
WPNJUJOH IFQBUJD MJWFS
QBJOBOENBMBJTF UJSFEOFTT
*UNBZUBLF JOBNNBUJPOBUUIFTJUFPGJOKFDUJPO
VQUPNPOUIGPSQBUJFOUTUPSFDPWFSBOETPNFQBUJFOUTNBZSFRVJSF
IPTQJUBMJTBUJPO:PVOHDIJMESFONBZOPUTIPXBOZTZNQUPNTCVUBSF
TUJMMJOGFDUJPVT1BUJFOUTBSFJOGFDUJPVTVQUPXFFLTCFGPSFUIFPOTFU
PGKBVOEJDFBOEGPSBQQSPYJNBUFMZXFFLBGUFSUIFKBVOEJDFBQQFBST

)FQBUJUJT#oWJSVTTQSFBENBJOMZCZCMPPE TFYVBMDPOUBDUPSGSPN "CPVUJODISPOJDDBSSJFSTXJMMEFWFMPQDJSSIPTJTPSMJWFSDBODFS


NPUIFSUPOFXCPSOCBCZDBVTFTBDVUFIFQBUJUJTPSDISPOJDDBSSJBHF

"CPVUJOXJMMIBWFJOKFDUJPOTJUFQBJOBOEJOXJMM
IBWFGFWFS"OBQIZMBYJTPDDVSTJOBCPVUJO

)JCoDPOUBHJPVTCBDUFSJBTQSFBECZSFTQJSBUPSZ
ESPQMFUTDBVTFTNFOJOHJUJT FQJHMPUUJUJT SFTQJSBUPSZ
PCTUSVDUJPO
TFQUJDBFNJBBOEPTUFPNZFMJUJT

"CPVUJONFOJOHJUJTQBUJFOUTEJFTBOEBCPVU
JOTVSWJWPSTIBTQFSNBOFOUCSBJOPSOFSWFEBNBHF
"CPVUJOFQJHMPUUJUJTQBUJFOUTEJF

"CPVUJOIBTEJTDPNGPSUPSMPDBM
JOBNNBUJPO"CPVUJOIBTGFWFS

)VNBOQBQJMMPNBWJSVToWJSVTTQSFBENBJOMZWJBTFYVBMDPOUBDU

"CPVUJOPGDFSWJDBMDBODFSTXPSMEXJEFIBWFCFFO
BTTPDJBUFEXJUI)17BOEJOXJUI)17

"CPVUJOXJMMIBWFQBJOBOEJOXJMMIBWF
TXFMMJOHSFEOFTTBUUIFTJUFPGJOKFDUJPO7FSZPDDBTJPOBMMZ
IFBEBDIF GFWFSBOEOBVTFBNBZPDDVS

*OVFO[BoDPOUBHJPVTWJSVTTQSFBECZSFTQJSBUPSZESPQMFUT
DBVTFTGFWFS NVTDMFBOEKPJOUQBJOT QOFVNPOJB

$BVTFTJODSFBTFEIPTQJUBMJTBUJPOJOUIFFMEFSMZ)JHISJTL
HSPVQTJODMVEFUIFFMEFSMZ EJBCFUJDTBOEBMDPIPMJDT

"CPVUJOIBTMPDBMSFBDUJPOT(VJMMBJO#BSS
TZOESPNFPDDVSTJOBCPVUJONJMMJPO

.FBTMFToIJHIMZJOGFDUJPVTWJSVTTQSFBECZ
ESPQMFUTDBVTFTGFWFS DPVHIBOESBTI

JODIJMESFOXJUINFBTMFTEFWFMPQTQOFVNPOJBBOE
JOEFWFMPQTFODFQIBMJUJT CSBJOJOBNNBUJPO
'PS
FWFSZDIJMESFOXIPEFWFMPQNFBTMFTFODFQIBMJUJT EJFT
BOEIBWFQFSNBOFOUCSBJOEBNBHF"CPVUJO
EFWFMPQT441& CSBJOEFHFOFSBUJPO
XIJDIJTBMXBZTGBUBM

"CPVUJOIBTEJTDPNGPSU MPDBMJOBNNBUJPOPSGFWFS"CPVUJO
EFWFMPQTBSBTIXIJDIJTOPOJOGFDUJPVT'FXFSUIBOJONJMMJPO
SFDJQJFOUTNBZEFWFMPQFODFQIBMJUJT JOBNNBUJPOPGUIFCSBJO


.FOJOHPDPDDBMJOGFDUJPOToCBDUFSJBTQSFBECZSFTQJSBUPSZ
ESPQMFUT$BVTFTFQUJDBFNJB JOGFDUJPOPGUIFCMPPETUSFBN
BOE
NFOJOHJUJT JOGFDUJPOPGUIFUJTTVFTTVSSPVOEJOHUIFCSBJO


"CPVUJOQBUJFOUTEJFT0GUIPTFUIBUTVSWJWF JOIBTTFWFSF $POKVHBUFWBDDJOF"CPVUJOIBTMPDBMJOBNNBUJPO 


TLJOTDBSSJOHPSMPTTPGMJNCT BOEJOIBTTFWFSFCSBJOEBNBHF GFWFS JSSJUBCJMJUZ BOPSFYJBPSIFBEBDIFT

.VNQToDPOUBHJPVTWJSVTTQSFBECZTBMJWBDBVTFT
TXPMMFOOFDLBOETBMJWBSZHMBOETBOEGFWFS

JODIJMESFOEFWFMPQTFODFQIBMJUJTJONBMFT
QBTUQVCFSUZEFWFMPQJOBNNBUJPOPGUIFUFTUFT
0DDBTJPOBMMZ NVNQTDBVTFTJOGFSUJMJUZPSEFBGOFTT

JOWBDDJOFSFDJQJFOUTNBZEFWFMPQTXFMMJOHPGUIFTBMJWBSZ
HMBOETJONJMMJPOSFDJQJFOUTEFWFMPQTNJMEFODFQIBMJUJT

1FSUVTTJToDPOUBHJPVTCBDUFSJBTQSFBECZSFTQJSBUPSZESPQMFUT
DBVTFTXIPPQJOHDPVHIBOEWPNJUJOHMBTUJOHVQUPNPOUIT

"CPVUJOXIPPQJOHDPVHIQBUJFOUTVOEFSUIFBHF
PGNPOUITEJFTGSPNQOFVNPOJBPSCSBJOEBNBHF

%51BE5QBWBDDJOFoBCPVUJOIBTMPDBMJOBNNBUJPOPS
GFWFS#PPTUFSEPTFTPG%51BNBZPDDBTJPOBMMZCFBTTPDJBUFEXJUI
FYUFOTJWFDJSDVNGFSFOUJBMTXFMMJOHPGUIFMJNC CVUUIJTSFTPMWFT
DPNQMFUFMZXJUIJOBGFXEBZT4FSJPVTBEWFSTFFWFOUTBSFWFSZSBSF

1OFVNPDPDDBMJOGFDUJPOToCBDUFSJBTQSFBECZSFTQJSBUPSZESPQMFUT "CPVUJONFOJOHJUJTQBUJFOUTEJFT
DBVTFTTFQUJDBFNJB NFOJOHJUJTBOEPDDBTJPOBMMZPUIFSJOGFDUJPOT

W1$7oBCPVUJOIBTMPDBMSFBDUJPOPSGFWFS

1PMJPoDPOUBHJPVTWJSVTTQSFBECZGBFDFTBOETBMJWBDBVTFT
GFWFS IFBEBDIFBOEWPNJUJOHBOENBZQSPHSFTTUPQBSBMZTJT

8IJMFNBOZJOGFDUJPOTDBVTFOPTZNQUPNT BCPVU
JOIPTQJUBMJTFEQBUJFOUTEJFTBOEJOQBUJFOUT
XIPTVSWJWFJTQFSNBOFOUMZQBSBMZTFE

-PDBMSFEOFTT QBJOBOETXFMMJOHBUUIFTJUFPGJOKFDUJPOBSF
DPNNPO6QUPJOIBTGFWFS DSZJOH BOEEFDSFBTFEBQQFUJUF

3PUBWJSVToWJSVTTQSFBECZGBFDBMPSBMSPVUF
DBVTFTHBTUSPFOUFSJUJTXIJDIDBOCFTFWFSF

*ODIJMESFOZFBSTPGBHF SPUBWJSVTJOGFDUJPOTJO"VTUSBMJB
BDDPVOUGPSBQQSPYJNBUFMZIPTQJUBMJTBUJPOTFWFSZZFBS 
BQQSPYJNBUFMZDIJMESFOWJTJUB(1BOEBQQSPYJNBUFMZ
DIJMESFOSFRVJSFBO&NFSHFODZ%FQBSUNFOUWJTJU*MMOFTT
NBZSBOHFGSPNNJME XBUFSZEJBSSIPFBPGMJNJUFEEVSBUJPOUP
TFWFSFEFIZESBUJOHEJBSSIPFBBOEGFWFSXIJDIDBOSFTVMUJOEFBUI

oJOWBDDJOFSFDJQJFOUTNBZEFWFMPQEJBSSIPFBPS
WPNJUJOHJOUIFXFFLGPMMPXJOHWBDDJOFBENJOJTUSBUJPO

3VCFMMBoDPOUBHJPVTWJSVTTQSFBECZESPQMFUTDBVTFT
GFWFS SBTIBOETXPMMFOHMBOET CVUDBVTFTTFWFSF
NBMGPSNBUJPOTJOCBCJFTPGJOGFDUFEQSFHOBOUXPNFO

"CPVUJOQBUJFOUTEFWFMPQBSBTIBOEQBJOGVMTXPMMFOHMBOET
JOBEPMFTDFOUTBOEBEVMUTIBWFQBJOGVMKPJOUTJO
EFWFMPQTUISPNCPDZUPQFOJB CSVJTJOHPSCMFFEJOH
JO
EFWFMPQTJOBNNBUJPOPGUIFCSBJOJOCBCJFTJOGFDUFEEVSJOH
UIFSTUXFFLTBGUFSDPODFQUJPOXJMMIBWFBNBKPSDPOHFOJUBM
BCOPSNBMJUZ JODMVEJOHEFBGOFTT CMJOEOFTTPSIFBSUEFGFDUT


"CPVUJOIBTEJTDPNGPSU MPDBMJOBNNBUJPO PS


GFWFS"CPVUJOIBTTXPMMFOHMBOET TUJOFDL PSKPJOU
QBJOT"CPVUJOIBTBSBTI XIJDIJTOPOJOGFDUJPVT
5ISPNCPDZUPQFOJB CSVJTJOHPSCMFFEJOH
PDDVSTBGUFS
BSTUEPTFPG..3BUBSBUFPGBCPVUJO

5FUBOVToDBVTFECZUPYJOPGCBDUFSJBJOTPJMDBVTFT
QBJOGVMNVTDMFTQBTNT DPOWVMTJPOT MPDLKBX

"CPVUJOQBUJFOUTEJFT5IFSJTLJT
HSFBUFTUGPSUIFWFSZZPVOHPSPME

%51BE5QBWBDDJOFoBCPVUJOIBTMPDBMJOBNNBUJPOPS
GFWFS#PPTUFSEPTFTPG%51BNBZPDDBTJPOBMMZCFBTTPDJBUFEXJUI
FYUFOTJWFDJSDVNGFSFOUJBMTXFMMJOHPGUIFMJNC CVUUIJTSFTPMWFT
DPNQMFUFMZXJUIJOBGFXEBZT4FSJPVTBEWFSTFFWFOUTBSFWFSZSBSF

7BSJDFMMB DIJDLFOQPY
oIJHIMZDPOUBHJPVTWJSVTDBVTFT
MPXHSBEFGFWFSBOEWFTJDVMBSSBTI3FBDUJWBUJPOPGUIF
WJSVTMBUFSJOMJGFDBVTFTIFSQFT[PTUFS TIJOHMFT


JOQBUJFOUTEFWFMPQFODFQIBMJUJT CSBJOJOBNNBUJPO

"CPVUJOQBUJFOUTEJF*OGFDUJPOEVSJOHQSFHOBODZDBO
SFTVMUJODPOHFOJUBMNBMGPSNBUJPOTJOUIFCBCZ0OTFUPGJOGFDUJPO
JOUIFNPUIFSGSPNEBZTCFGPSFUPEBZTBGUFSEFMJWFSZSFTVMUTJO
TFWFSFJOGFDUJPOJOUIFOFXCPSOCBCZJOVQUPPOFUIJSEPGDBTFT

"CPVUJOIBTBMPDBMSFBDUJPOPSGFWFS"NJMEWBSJDFMMB
MJLFSBTINBZEFWFMPQJOoJOSFDJQJFOUT

W117oBCPVUJOIBTBMPDBMSFBDUJPO

ADVERSE EVENTS FOLLOWING IMMUNISATION


Commonly observed adverse events following immunisation with vaccines used in the
National Immunisation Program (NIP) schedule and what to do about them
The following information can be photocopied and given as post-vaccination advice.
All the common adverse events following immunisation are usually mild and transient and treatment is not usually required. If the adverse event following
immunisation is severe or persistent, or if you are worried about yourself or your childs condition, see your doctor or immunisation clinic nurse as soon as possible
or go to a hospital. Adverse events may be reported via ADRAC, State and Territory Health Authorities or via immunisation service providers.
Diphtheria-tetanus-pertussis (acellular)
DTPa-containing vaccines and dTpa
(adolescent/adult) vaccines

Haemophilus influenzae type b vaccine (Hib)

Hepatitis A vaccine (HepA) (Indigenous


children NT, QLD, SA, WA)

Hepatitis B vaccine (HepB)

t -PDBMJTFEQBJO SFEOFTTBOE
swelling at injection site

t -PDBMJTFEQBJO SFEOFTTBOE
swelling at injection site

t -PDBMJTFEQBJO SFEOFTTBOE
swelling at injection site

t -PDBMJTFEQBJO SFEOFTTBOE
swelling at injection site

t 0DDBTJPOBMMZJOKFDUJPOTJUFOPEVMF NBZ
last many weeks (no treatment needed)

t 0DDBTJPOBMMZJOKFDUJPOTJUFOPEVMF NBZ
last many weeks (no treatment needed)

t -PXHSBEFUFNQFSBUVSF GFWFS

t 0DDBTJPOBMMZJOKFDUJPOTJUFOPEVMF NBZ
last many weeks (no treatment needed)

t -PXHSBEFUFNQFSBUVSF GFWFS

t -PXHSBEFUFNQFSBUVSF GFWFS

t -PXHSBEFUFNQFSBUVSF GFWFS

In children the following may also occur:


t *SSJUBCMF DSZJOH VOTFUUMFEBOE
generally unhappy
t %SPXTJOFTTPSUJSFEOFTT
Human papillomavirus vaccine (HPV)

Influenza vaccine

Measles-mumps-rubella vaccine (MMR)

Meningococcal C conjugate vaccine (MenCCV)

t -PDBMJTFEQBJO SFEOFTTBOE
swelling at injection site

t %SPXTJOFTTPSUJSFEOFTT

t 0DDBTJPOBMMZJOKFDUJPOTJUFOPEVMF NBZ
last many weeks (no treatment needed)

t *SSJUBCMF DSZJOH VOTFUUMFEBOE


generally unhappy

Seen 710 days after vaccination:

t -PTTPGBQQFUJUF

t 5FNQFSBUVSF GFWFS DBOCF0C)


MBTUJOHoEBZT GBJOUSFESBTI OPU
infectious), head cold and/or runny
nose, cough and/or puffy eyes

t )FBEBDIF VTVBMMZPCTFSWFE
in adolescents/adults)

t %SPXTJOFTTPSUJSFEOFTT

t 0DDBTJPOBMMZJOKFDUJPOTJUFOPEVMF NBZ
last many weeks (no treatment needed)

t -PXHSBEFUFNQFSBUVSF GFWFS

t .JMEIFBEBDIF
t .JMEOBVTFB

t .VTDMFBDIFT
t -PDBMJTFEQBJO SFEOFTTBOE
swelling at injection site
t 0DDBTJPOBMMZJOKFDUJPOTJUFOPEVMF NBZ
last many weeks (no treatment needed)
t -PXHSBEFUFNQFSBUVSF GFWFS

t 4XFMMJOHPGTBMJWBSZHMBOET

t -PDBMJTFEQBJO SFEOFTTBOE
swelling at injection site

t -PXHSBEFUFNQFSBUVSF GFWFS

Pneumococcal vaccines (conjugate


7vPCV and polysaccharide 23vPPV)

Inactivated poliomyelitis vaccine


(IPV) and IPV-containing vaccines

Rotavirus vaccine

Varicella vaccine (VV)

t -PDBMJTFEQBJO SFEOFTTBOE
swelling at injection site

t .VTDMFBDIFT

t 7PNJUJOHBOEEJBSSIPFBDBOPDDVSVQ
to 7 days following vaccination

t -PDBMJTFEQBJO SFEOFTTBOE
swelling at injection site

t 0DDBTJPOBMMZJOKFDUJPOTJUFOPEVMF NBZ
last many weeks (no treatment needed)
t -PXHSBEFUFNQFSBUVSF GFWFS

t -PDBMJTFEQBJO SFEOFTTBOE
swelling at injection site

t 0DDBTJPOBMMZJOKFDUJPOTJUFOPEVMF NBZ
last many weeks (no treatment needed)

t 0DDBTJPOBMMZJOKFDUJPOTJUFOPEVMF NBZ
last many weeks (no treatment needed)

t 5FNQFSBUVSF GFWFS DBOCF0C)

t -PXHSBEFUFNQFSBUVSF GFWFS

Seen 526 days after vaccination:


t 1VTUVMBSSBTI oMFTJPOT
VTVBMMZBUJOKFDUJPO
site, occasionally other parts of the body

Key to table
DTPa

diphtheria-tetanus-pertussis acellular (infant/child formulation)

IPV

inactivated poliomyelitis vaccine

dTpa

diphtheria-tetanus-pertussis acellular (adolescent/adult formulation)

MenCCV

meningococcal C conjugate vaccine

HepA

hepatitis A vaccine

MMR

measles-mumps-rubella vaccine

HepB

hepatitis B vaccine

7vPCV

7-valent pneumococcal conjugate vaccine

Hib

Haemophilus influenzaeUZQFCWBDDJOF 1310.1PS1315

23vPPV

WBMFOUQOFVNPDPDDBMQPMZTBDDIBSJEFWBDDJOF

HPV

human papillomavirus vaccine

Rotavirus

rotavirus vaccine

Influenza

influenza or flu vaccine

VV

varicella vaccine

What to do to manage injection site discomfort


.BOZWBDDJOFJOKFDUJPOTNBZSFTVMUJOTPSFOFTT SFEOFTT JUDIJOH TXFMMJOHPSCVSOJOHBUUIFJOKFDUJPOTJUFGPSUPEBZT1BSBDFUBNPMNJHIUCFSFRVJSFEUPFBTFUIF
EJTDPNGPSU4PNFUJNFTBTNBMM IBSEMVNQNBZQFSTJTUGPSTPNFXFFLTPSNPOUIT5IJTTIPVMEOPUCFPGDPODFSOBOESFRVJSFTOPUSFBUNFOU
Managing fever after immunisation
(JWFFYUSBVJETUPESJOL%POPUPWFSESFTTBOJOGBOUJGIPU"MUIPVHIUIFSPVUJOFVTFPGQBSBDFUBNPMBUUIFUJNFPGWBDDJOBUJPOJTOPMPOHFSOFDFTTBSZ JUNBZCFSFRVJSFEJG GPSFYBNQMF 
BOJOGBOUPSDIJMEIBTBIJHIGFWFSGPMMPXJOHWBDDJOBUJPO5IFEPTFPGQBSBDFUBNPMJTNHLHPGQBSBDFUBNPMMJRVJE VQUPBNBYJNVNEBJMZEPTFPGNHLHQFSEBZ

Copyright
Australian Government 2008
Paper-based publication
This work is copyright. Apart from any use permitted under the Copyright Act 1968, no part may
be reproduced by any process without written permission from the Commonwealth. Requests
and inquiries concerning reproduction and rights should be addressed to the Commonwealth
Copyright Administration, Attorney-Generals Department, Robert Garran Offices, National
Circuit, Canberra, ACT, 2600 or posted at: http://www.ag.gov.au/cca
ISBN 1-74186-483-6
Electronic documents
This work is copyright. You may download, display, print and reproduce this material in unaltered
form only (retaining this notice) for your personal, non-commercial use, or use within your
organisation. Apart from any use as permitted under the Copyright Act 1968, all other rights are
reserved. Requests for further authorisation should be directed to the Commonwealth Copyright
Administration, Attorney-Generals Department, Robert Garran Offices, National Circuit,
Canberra, ACT, 2600 or posted at: http://www.ag.gov.au/cca
Online ISBN: 1-74186-484-4
Publication Approval Number: 2923

Disclaimer
While every effort has been made to check drug dosage recommendations in this Handbook, it is
still possible that errors have been missed. Furthermore, dosage recommendations are continually
being revised and new adverse events recognised.
Trade names used in this publication are for identification purposes only. Their use does not
imply endorsement of any particular brand of drug or vaccine. This Handbook is a general guide
to appropriate practice subject to clinicians judgement in each individual case. It is designed
to provide information to assist decision making using the best information available at date
of National Health and Medical Research Council approval (11 October 2007). The Australian
Government Department of Health and Ageing does not accept any liability for any injury, loss or
damage incurred by use of or reliance on the information.

The NHMRC
The National Health and Medical Research Council (NHMRC) is Australias leading funding body
for health and medical research. The NHMRC also provides the government, health professionals
and the community with expert and independent advice on a range of issues that directly affect
the health and well being of all Australians.
The NHMRC provided support to this project through its Guidelines Assessment Register (GAR)
process. The GAR consultant on this project was Biotext Pty Ltd. These Guidelines, apart from
Chapters 3.7 and 3.9, were approved by the Chief Executive Officer of the NHMRC under Section
14A of the National Health and Medical Research Council Act, 1992 on 17 July 2007. The remaining
two chapters were approved on 11 October 2007.

The ATAGI
The Australian Technical Advisory Group on Immunisation (ATAGI) was established by the then
Minister for Health and Family Services in 1998 to provide expert technical and scientific advice on
the Immunise Australia Program and to work cooperatively with the NHMRC on issues such as the
The Australian Immunisation Handbook.

The Handbook
This Handbook is published approximately every three years but changes to the recommendations or
schedule may occur between publications. The Handbook and any changes between publications are
available on the website: www.immunise.health.gov.au. This hardcopy version of the Handbook does
not contain any references these are available on the electronic version.
Ninth Edition January 2008

ii The Australian Immunisation Handbook 9th Edition

PREFACE
The 9th edition of The Australian Immunisation Handbook was prepared by the
Australian Technical Advisory Group on Immunisation of the Australian Government
Department of Health and Ageing.

Members of the Australian Technical


Advisory Group on Immunisation
Chair
Professor Terry Nolan, Paediatrician and Epidemiologist and Head, School of
Population Health, The University of Melbourne, VIC.

Members
Ms Jenny Bourne, Assistant Secretary, Targeted Prevention Programs Branch,
Population Health Division, Australian Government Department of Health and
Ageing, ACT.
Ms Sue Campbell-Lloyd, Manager, Immunisation Unit, AIDS/Infectious Diseases
Branch, NSW Health, NSW.
Dr Grahame Dickson, Medical Officer, Drug Safety and Evaluation Branch,
Therapeutic Goods Administration, Australian Government Department of Health
and Ageing, ACT.
Dr Nicole Gilroy, Staff Specialist, Infectious Diseases, Westmead Hospital, Centre
for Infectious Diseases and Microbiology, Western Sydney Area Health Service, and
Infectious Diseases Physician, BMT Network, NSW.
Dr Jeffrey Hanna, Medical Director, Communicable Disease Control, Tropical
Population Health Unit, Queensland Health, QLD.
Ms Jenni Howlett, State President, Child Health Association, TAS.
Clinical Professor David Isaacs, Paediatrician, Department of Immunology and
Infectious Diseases, The Childrens Hospital at Westmead, NSW.
Ms Ann Kempe, Surveillance Manager, CCRE in Child and Adolescent Immunisation,
Murdoch Childrens Research Institute, Royal Childrens Hospital, VIC.
Dr Rosemary Lester, Assistant Director, Public Health Branch, Communicable Disease
Control Unit, Department of Human Services, VIC.
Professor Peter McIntyre, Professor of Paediatrics and Preventive Medicine, The
University of Sydney, and Director, National Centre for Immunisation Research and
Surveillance of Vaccine Preventable Diseases, The University of Sydney and The
Childrens Hospital at Westmead, NSW.
Dr Joanne Molloy, General Practitioner, Medical Officer of Health, City of Greater
Geelong, and Immunisation Program Manager, GP Association of Geelong, VIC.
Associate Professor Michael Nissen, Director of Infectious Diseases and Clinical
Microbiologist, Unit Head of Queensland Paediatric Infectious Disease Laboratory,
and Associate Professor in Biomolecular, Biomedical Science and Health, Royal
Childrens Hospital, QLD.

Preface iii

Dr Rod Pearce, General Practitioner, Medical Officer of Health, Eastern Health


Authority, Adelaide and GP Immunisation Advisor, Adelaide Central and Eastern
Division of General Practice, SA.
Dr Peter Richmond, Senior Lecturer, University of Western Australia, School of
Paediatrics and Child Health, and General Paediatrician and Paediatric Immunologist,
Princess Margaret Hospital for Children, WA.
Dr Sue Skull, Paediatrician, Clinical Epidemiologist and Public Health Physician, and
Senior Lecturer, Department of Paediatrics, The University of Melbourne, VIC.

Secretary
Ms Letitia Toms, Director, Immunisation Policy Section, Targeted Prevention Programs
Branch, Population Health Division, Australian Government Department of Health
and Ageing, ACT.

Secretariat support, Australian Technical


Advisory Group on Immunisation
Ms Brigid Dohnt, Mrs Claire Kellie, Ms Jacinta Holdway, Mr John Mohoric,
Ms Sally Warild.

Technical Editors
Dr Jane Jelfs, Immunisation Handbook and Policy Support Coordinator, National
Centre for Immunisation Research and Surveillance of Vaccine Preventable Diseases,
The University of Sydney and The Childrens Hospital at Westmead, NSW.
Dr Kristine Macartney, Senior Research Fellow, National Centre for Immunisation
Research and Surveillance of Vaccine Preventable Diseases, The University of Sydney
and The Childrens Hospital at Westmead, NSW.

Handbook Technical Support


Ms Catherine King, Information Manager, National Centre for Immunisation Research
and Surveillance of Vaccine Preventable Diseases, The University of Sydney and The
Childrens Hospital at Westmead, NSW.
Ms Donna Armstrong, Communications Officer, National Centre for Immunisation
Research and Surveillance of Vaccine Preventable Diseases, The University of Sydney
and The Childrens Hospital at Westmead, NSW.

Technical Writers
Dr Julia Brotherton, Dr Tony Gherardin, Ms Heather Gidding, Dr Kate Hale, Dr Jeffrey
Hanna, Ms Anita Heywood, Professor David Isaacs, Dr Jane Jelfs, Ms Ann Kempe,
DrKristine Macartney, Professor Peter McIntyre, Mr Robert Menzies, Dr Joanne
Molloy, Dr Helen Quinn, Dr Yashwant Sinha, Dr Nicholas Wood.

iv The Australian Immunisation Handbook 9th Edition

Acknowledgments
Associate Professor Ross Andrews

Dr Amanda Leach

Professor Graeme Barnes

Dr Julie Leask

Professor Richard Benn

Professor Raina MacIntyre

Associate Professor Beverley Biggs

Professor Barrie Marmion

Professor Julie Bines

Dr Helen Marshall

Dr Ian Boyd

Dr Brad McCall

Professor Anthony Brown

Dr Treasure McGuire

Professor Margaret Burgess

Ms Cathlyn McInnes

Dr Jim Buttery

Dr Moira McKinnon

Professor Jonathan Carapetis

Dr Jodie McVernon

Dr Louise Causer

Dr Ann Mijch

Ms Patricia Coward

Professor Kim Mulholland

Dr Angela Dean

Dr Neil Parker

Dr Ki Douglas

Ms Karen Peterson

Ms Barbara Eldred

Ms Susie Prest

Mr Lloyd Ellis

Professor William Rawlinson

Professor Kevin Forsyth

Dr Jenny Royle

Professor Lyn Gilbert

Dr Tilman Ruff

Dr Mike Gold

Ms Andrea Schaffer

Dr Robert Hall

Dr Rosalie Schultz

Dr Alan Hampson

Dr Christine Selvey

Professor Mark Harris

Dr Linda Selvey

Ms Trish Harris

Ms Barbara Steadman

Dr Bronwen Harvey

Dr John Sullivan

Dr Bob Kass

Mr Sean Tarrant

Dr Heath Kelly

Dr Diana Thomas

Dr Vicki Krause

Dr Bruce Thorley

Dr Andrew Langley

Dr Melanie Wong

Dr Glenda Lawrence

Dr Margaret Young

Preface v

Contents
PART 1: VACCINATION PROCEDURES 

Introduction to The Australian Immunisation Handbook

1.1 Whats new?

1.2 An overview of vaccination preface to Chapters 1.31.5

1.3 Pre-vaccination procedures


1.3.1 Preparing an anaphylaxis response kit
1.3.2 Effective cold chain: transport, storage and handling of vaccines
1.3.3 Valid consent
1.3.4 Pre-vaccination screening
1.3.5 Catch-up

8
8
8
12
14
21

1.4 Administration of vaccines


1.4.1 Occupational health and safety issues
1.4.2 Equipment for vaccination
1.4.3 Route of administration
1.4.4 Preparation for vaccine administration
1.4.5 Vaccine injection techniques
1.4.6 Recommended injection sites
1.4.7 Positioning for vaccination
1.4.8 Identifying the injection site
1.4.9 Administering multiple vaccine injections at the same visit

39
39
39
41
43
44
45
47
51
56

1.5 Post-vaccination procedures


1.5.1 Immediate after-care
1.5.2 Adverse events following immunisation
1.5.3 Documentation of vaccination
1.5.4 The Australian Childhood Immunisation Register

58
58
58
66
67

PART 2: Vaccination for special risk groups

70

2.1 Vaccination for Aboriginal and Torres Strait Islander people

70

2.2 Vaccination for international travel 

75

2.3 Groups with special vaccination requirements


2.3.1 Vaccination of children who have had a serious adverse event
following immunisation (AEFI)
2.3.2 Vaccination of women planning pregnancy, pregnant or
breastfeeding women, and preterm infants 
2.3.3 Vaccination of individuals with impaired immunity due to disease
or treatment
2.3.4 Vaccination of recent recipients of normal human immunoglobulin
2.3.5 Vaccination of patients following receipt of other blood products
including blood transfusions
2.3.6 Vaccination of patients with bleeding disorders
2.3.7 Vaccination before or after anaesthesia/surgery
2.3.8 Vaccination of those at occupational risk

84

vi The Australian Immunisation Handbook 9th Edition

84
84
90
102
102
104
104
104

2.3.9
2.3.10
2.3.11
2.3.12

Vaccination of immigrants to Australia


Vaccination of inmates of correctional facilities 
Vaccination of men who have sex with men 
Vaccination of injecting drug users 

PART 3: Vaccines listed by disease

108
108
108
109

110

3.1 Australian bat lyssavirus infection and rabies 

110

3.2 Cholera

120

3.3 Diphtheria

124

3.4 Haemophilus influenzae type b (Hib)

131

3.5 Hepatitis A 

139

3.6 Hepatitis B

149

3.7 Human papillomavirus

164

3.8 Immunoglobulin preparations 

175

3.9 Influenza 

184

3.10 Japanese encephalitis 

195

3.11 Measles

201

3.12 Meningococcal disease

213

3.13 Mumps 

223

3.14 Pertussis

227

3.15 Pneumococcal disease

240

3.16 Poliomyelitis

251

3.17 Q fever 

257

3.18 Rotavirus 

265

3.19 Rubella 

274

3.20 Smallpox

283

3.21 Tetanus

288

3.22 Tuberculosis

297

3.23 Typhoid

303

3.24 Varicella

309

3.25 Yellow fever 

322

3.26 Zoster (herpes zoster)

329

Contents vii

Appendix 1: Contact details for Australian, State and Territory Government


health authorities and communicable disease control

332

Appendix 2: Handbook development

334

Appendix 3: Products registered in Australia but not currently available 

339

Appendix 4: Components of vaccines used in the National Immunisation


Program

340

Appendix 5: Commonly asked questions about vaccination

344

Appendix 6: Definitions of adverse events following immunisation

360

Appendix 7: Glossary of technical terms 

364

Appendix 8: List of commonly used abbreviations

369

Appendix 9: Dates when vaccines became available in Australia 

372

Appendix 10: Summary table procedures for a vaccination encounter

375

Index

378

viii The Australian Immunisation Handbook 9th Edition

Index of Tables
Table 1.3.1: Pre-vaccination screening checklist

16

Table 1.3.2: Responses to relevant conditions or circumstances identified


by the pre-vaccination screening checklist

17

Table 1.3.3: Live attenuated parenteral and oral vaccines

20

Table 1.3.4: False contraindications to vaccination

21

Table 1.3.5: Number of vaccine doses that should have been administered
by the current age of the child (table to be used in conjunction
with Catch-up Worksheet)

28

Table 1.3.6: Minimum dose intervals for NIP vaccines for children <8years
of age (table to be used in conjunction with Catch-up Worksheet)

29

Table 1.3.7: Minimum age for the first dose of vaccine in exceptional
circumstances

30

Table 1.3.8: Recommendations for Hib catch-up vaccination for children


<5years of age when doses have been delayed or missed

33

Table 1.3.9: Recommendations for pneumococcal catch-up vaccination for


low-risk children (including Indigenous children living in ACT,
NSW, VIC and TAS) <2years of age, when doses have been
delayed or missed

34

Table 1.3.10: Recommendations for pneumococcal catch-up vaccination for


Indigenous children <2years of age in NT, QLD, SA and WA,
when doses have been delayed or missed

35

Table 1.3.11: Recommendations for pneumococcal catch-up vaccination for


children 5years of age with underlying medical conditions

36

Table 1.3.12: Catch-up schedules for individuals 8years of age

38

Table 1.4.1: Route of administration for vaccines commonly used in


Australia

42

Table 1.4.2: Recommended needle size, length and angle for administering
vaccines

45

Table 1.5.1: Clinical features which may assist differentiation between a


vasovagal episode and anaphylaxis

62

Table 1.5.2: Doses of intramuscular 1:1000 (one in one thousand)


adrenaline for anaphylaxis

64

Table 1.5.3: Contact details for notification of AEFI

66

Table 2.2.1: Dose and routes of administration of commonly used vaccines


in adult travellers (15years of age)

80

Table 2.2.2: Recommended lower age limits of travel vaccines for children

82

Table 2.3.1: Vaccinations in pregnancy

86

Table 2.3.2: Recommendations for vaccinations for solid organ transplant


(SOT) recipients

95

Contents ix

Table 2.3.3: Post-transplantation vaccination schedules for allogeneic and


autologous haematopoietic stem cell transplant recipients

98

Table 2.3.4: Immunological categories based on age-specific CD4 counts


and percentage of total lymphocytes

99

Table 2.3.5: Recommended intervals between either immunoglobulins or


blood products and MMR, MMRV or varicella vaccination

103

Table 2.3.6: Recommended vaccinations for those at risk of occupationally


acquired vaccine-preventable diseases

105

Table 3.1.1: Summary of Australian bat lyssavirus and rabies postexposure treatment for non-immune individuals

117

Table 3.5.1: Recommended dosages and schedules for use of the


inactivated hepatitis A vaccines

143

Table 3.5.2: Recommended doses of normal human immunoglobulin


(NHIG) to be given as a single intramuscular injection to close
contacts of hepatitis A cases

146

Table 3.6.1: Hepatitis B and hepatitis A/hepatitis B combination


vaccination schedules 

155

Table 3.6.2: Accelerated hepatitis B vaccination schedules

156

Table 3.6.3: Post-exposure prophylaxis for non-immune individuals


exposed to an HBsAg positive person

162

Table 3.9.1: Recommended doses of influenza vaccine

189

Table 3.11.1: Management of significant measles exposure using


vaccination or normal human immunoglobulin (NHIG)

212

Table 3.12.1: Early clinical management of suspected meningococcal disease

220

Table 3.14.1: Recommended antimicrobial therapy and chemoprophylaxis


regimens for pertussis in infants, children and adults

236

Table 3.15.1: Summary table pneumococcal vaccination schedule for


children 9years of age (see also Section 1.3.5, Catch-up)

245

Table 3.15.2: Underlying medical conditions predisposing children 9years


of age to IPD

246

Table 3.15.3: Revaccination with 23vPPV for people 10years of age

248

Table 3.17.1: Interpretation and action for serological and skin test results

261

Table 3.18.1: Age limits for dosing of oral rotavirus vaccines

269

Table 3.21.1: Guide to tetanus prophylaxis in wound management

294

Table 3.24.1: Recommendations for varicella vaccination with monovalent


VV (currently available), and once MMRV vaccines are available

313

Table 3.24.2: Zoster immunoglobulin-VF (ZIG) dose based on weight

320

Table 3.25.1: Yellow fever endemic countries

324

x The Australian Immunisation Handbook 9th Edition

Index of Figures
Figure 1.3.1: Catch-up Worksheet for children <8years of age

27

Figure 1.4.1: The cuddle position for vaccination of a child <12months of age

47

Figure 1.4.2: Positioning an infant on an examination table for vaccination

48

Figure 1.4.3: Positioning an older child in the cuddle position

49

Figure 1.4.4: Positioning a child in the straddle position

50

Figure 1.4.5: Diagram of the muscles of the thigh showing the anatomical
markers to identify the recommended (vastus lateralis)
injection site

52

Figure 1.4.6: Photograph of the thigh showing the recommended (vastus


lateralis) injection site

52

Figure 1.4.7: Diagram showing the anatomical markers to identify the


ventrogluteal injection site

53

Figure 1.4.8: Photograph with infant prone across carers lap, showing
markers to identify the ventrogluteal injection site

54

Figure 1.4.9: Diagram showing the anatomical markers to identify the


deltoid injection site

55

Figure 1.4.10: A subcutaneous injection into the deltoid area of the upper arm
using a 25 gauge, 16mm needle, inserted at a 45 angle

55

Figure 1.4.11: Recommended technique for giving multiple vaccine injections


to an infant <12months of age into the anterolateral thigh

56

Figure 3.4.1:

 aemophilus influenzae type b (Hib) notifications, presumed


H
Hib hospitalisations and deaths of children aged 0 to 4years
from Hib, Australia 1993 to 2005

133

Figure 3.5.1: Notifications of hepatitis A in Australia, 1991 to 2006

140

Figure 3.6.1: The influence of age of infection with the hepatitis B virus on
the likelihood of becoming a hepatitis B carrier 

150

Figure 3.7.1: The dynamic relationship between HPV infection and


cervical health

167

Figure 3.9.1: Influenza notification rates 20032005 and hospitalisation


rates 2002/2003 to 2004/2005, Australia, by age group

186

Figure 3.10.1: Map of the Torres Strait

196

Figure 3.14.1: Pertussis notifications by year of onset, Australia 19932005

228

Figure 3.17.1: Q fever notifications and hospitalisations, Australia, 1993 to


2005, bymonth of diagnosis or admission

258

Contents xi

xii The Australian Immunisation Handbook 9th Edition

PART 1: VACCINATION PROCEDURES


Introduction to The Australian
Immunisation Handbook
For more than 200years, since Edward Jenner first demonstrated that vaccination
offered protection against smallpox, the use of vaccines has continued to
reduce the burden of many bacterial and viral diseases. As a result of successful
vaccination programs, deaths from tetanus, diphtheria, Haemophilus influenzae
type b and measles are now extremely rare in Australia.1
Vaccination not only protects individuals, but also others in the community, by
increasing the general level of immunity and minimising the spread of infection. It
is vital that healthcare professionals take every available opportunity to vaccinate
children and adults. It is also important that the public be made aware of the
proven effectiveness of immunisation to save lives and prevent serious illness.
The purpose of The Australian Immunisation Handbook is to provide clinical
guidelines for health professionals on the safest and most effective use of
vaccines in their practice. These recommendations are developed by the
Australian Technical Advisory Group on Immunisation (ATAGI) and endorsed
by the National Health and Medical Research Council (NHMRC).
The Handbook provides clinical recommendations based on the best scientific
evidence available at the time of publication from published and unpublished
literature. Further details regarding the Handbook revision procedures are
described in Appendix 2. Where specific empiric evidence was unavailable,
recommendations were formulated using the best available expert opinion
relevant to Australia. The reference lists for all chapters are included in the
electronic version of the Handbook which is available via the Immunise Australia
website (www.immunise.health.gov.au). The electronic version of the Handbook
has additional information regarding recommendations in the new vaccine
chapters, including systematic reviews of the literature.
In some instances, the NHMRC recommendations differ from vaccine product
information sheets (PI); these differences are detailed in the relevant vaccine
chapters under the heading Variations from product information. Where a
variation exists, the NHMRC recommendation should be considered best practice.
The information contained within the Handbook was correct at the time of
printing. However, the content of the Handbook is reviewed regularly. The 9th
edition of The Australian Immunisation Handbook will remain current unless
amended electronically via the Immunise Australia website or until the 10th
edition of the Handbook is published.

Introduction 1

ELECTRONIC UPDATES to the 9th edition of The Australian


Immunisation Handbook will be available at:
www.immunise.health.gov.au

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

2 The Australian Immunisation Handbook 9th Edition

1.1 Whats new?


All chapters have been updated and revised where necessary. The 9th edition
introduces new vaccines, changes to the schedules, changes to recommendations
and procedures regarding the administration of vaccines, and changes to the
presentation of the Handbook. Some changes made since the publication of the
hard copy of the 8th edition of the Handbook in September 2003, and before the
9th edition, were available online on the Immunise Australia website and are not
described below.
The term Australian Standard Vaccination Schedule (ASVS) is no longer used in
the Handbook.
The National Immunisation Program (NIP) is used throughout the Handbook and
refers to funded vaccines as they appear on the National Immunisation Program
(NIP) schedule. The NIP schedule may change over time and is available via the
Immunise Australia website (www.immunise.health.gov.au).

New chapters and chapters which no


longer appear in the Handbook
Three new chapters have been included in the Handbook 3.7 Human
papillomavirus, 3.18 Rotavirus and 3.26 Zoster.
There are 4 new appendices in the Handbook. These are Handbook
development (Appendix 2); a list of vaccines which are registered in
Australia but either not currently available or no longer available in Australia
(Appendix 3); a list of major components in the vaccines in the National
Immunisation Program (Appendix 4); and a table that is a summary of
procedures for a vaccination encounter (Appendix 10).
Two chapters have been deleted: anthrax and plague. For information about
anthrax, please go to the Australian Government website www.health.gov.au
and use the index option to select the anthrax fact sheet.
Three chapters previously in the 8th edition, botulism, cytomegalovirus and
respiratory syncytial virus, have been incorporated into the immunoglobulin
chapter (3.8 Immunoglobulin preparations).

Overview of major changes to existing


recommendations and procedures
Part 1
The layout of Part 1 has been altered from previous editions of the Handbook
into 3 chapters which are described in Chapter 1.2 An overview of vaccination.
The new layout applies to Chapter 1.3 Pre-vaccination procedures (including
cold chain, consent, pre-vaccination screening and catch-up); Chapter 1.4

What's new? 3

1.1 Whats new?

Changes introduced in this edition of the Handbook

Administration of vaccines (including route, needle size and injection site); and
Chapter 1.5 Post-vaccination procedures (including adverse events following
immunisation, and the recording of vaccinations).
Advice on preparing an anaphylaxis response kit has been added to prevaccination procedures, Chapter 1.3.
The pre-vaccination screening checklist and assessment table have been
revised and recommended steps for screening added to the section,
Chapter1.3.
The cold chain guidelines have been updated in Chapter 1.3 and the
recommendations summarised to reflect and reference the National Vaccine
Storage Guidelines: Strive for 5.
The valid consent section has been redrafted and updated.
The recommended anatomical site for intramuscular (IM) administration of
vaccines in infants <12months of age is the anterolateral thigh.
The recommended anatomical site for IM administration of vaccines in those
12months of age is the deltoid.
The ventrogluteal area is included as an alternative anatomical site for the
administration of vaccines at any age. This is based on published data. It is
important that vaccine providers using this site are trained in the recognition
of the relevant anatomical landmarks.
The recommended needle size and length for IM injection is 23 or 25 gauge,
25 mm in length.
The recommended angle of insertion of the needle for IM administration of
vaccines is 90 to the skin surface.
Injection techniques have been described in more detail with additional
photographs and/or diagrams demonstrating positions and the
recommended anatomical sites.
Catch-up schedules have been updated for vaccines in the National
Immunisation Program and practical tools to assist with catch-up added.
A table of catch-up schedules for individuals aged 8years has been included.
Information on reporting of adverse events following immunisation has been
updated to reflect recent changes to the national reporting arrangements.
Information on the Australian Childhood Immunisation Register has been
updated.
The table previously in the 8th edition Handbook entitled Information on
vaccines exposed to different temperatures has been deleted as some of the
information is no longer considered valid.
Management of anaphylaxis with 1:10000 adrenaline is no longer
recommended; use of 1:1000 adrenaline is recommended.

4 The Australian Immunisation Handbook 9th Edition

Tools to photocopy include the pre-vaccination checklist, catch-up work


sheet, and Appendix 10 Summary table procedures for a vaccination encounter.
Recommendations for groups with special vaccination requirements (Chapter
2.3) have undergone substantial revision and incorporate new tables and
separate sections for pregnant and breastfeeding women and women
planning pregnancy, preterm infants, people with impaired immunity,
oncology patients and transplant recipients.
Recommendations for immunisation of certain occupational groups have
been expanded (Chapter 2.3).
A comprehensive table outlining the suggested intervals between receipt
of either a blood product or an immunoglobulin-containing product and
administration of either measles, mumps, rubella or varicella vaccines is
included (Chapter 2.3).

Part 3
Chapters 3.3 Diphtheria and 3.21 Tetanus
For adults requiring a primary course of dT, dTpa is recommended for the
first dose followed by 2 doses of dT (or dTpa only if dT is unavailable).

Chapter 3.6 Hepatitis B


For preterm babies, recommendations for hepatitis B vaccination have been
revised.

Chapter 3.8 Immunoglobulin preparations


Botulism, cytomegalovirus and respiratory syncytial virus are now incorporated
into this chapter.

Chapter 3.9 Influenza


For children aged 6months to <3years the dose of influenza vaccine is 0.25mL.
It is recommended that all Aboriginal and Torres Strait Islander people aged
15years receive annual influenza vaccination.
It is recommended that children 6months of age and adults with a chronic
neurological condition receive annual influenza vaccination.

Chapters 3.11 Measles, 3.13 Mumps and 3.19 Rubella


The second dose of MMR vaccine is recommended at 18months of age, not at
4years of age.
The use of MMRV vaccines, when available, is discussed.

What's new? 5

1.1 Whats new?

Part 2

Chapter 3.12 Meningococcal disease


Close household contacts of a case of invasive meningococcal disease should
be vaccinated as well as receiving antibiotic prophylaxis.

Chapter 3.14 Pertussis


For adults requiring a primary course of dT, dTpa is recommended for the
first dose followed by 2 doses of dT (or dTpa only if dT is unavailable).
A new table detailing antibiotic prophylaxis for pertussis cases and their
contacts has been included.

Chapter 3.15 Pneumococcal disease


Children 9years of age with specified underlying medical conditions should
receive 2 doses of 7-valent pneumococcal conjugate vaccine followed by a
dose of 23-valent pneumococcal polysaccharide vaccine.
Recommendations for revaccination of adults with 23-valent pneumococcal
polysaccharide vaccine have been revised and tabulated.

Chapter 3.24 Varicella


When combination measles, mumps, rubella and varicella vaccine/s (MMRV)
become available, it is recommended that varicella vaccination be given at
12months of age, using MMRV.
Administration of a second dose of varicella-containing vaccine in children
aged <14years is recommended to minimise the risk of breakthrough disease.

Chapter 3.25 Yellow fever


Yellow fever vaccine is now recommended for travellers (provided there are
no contraindications) going to urban and/or rural areas of endemic countries.

6 The Australian Immunisation Handbook 9th Edition

1.2 An overview of vaccination


preface to Chapters 1.31.5
The following sections of Part 1 (Chapters 1.31.5) describe chronologically the
steps involved around a vaccination encounter, starting with pre-vaccination
requirements (Chapter 1.3), then administration of vaccines (Chapter 1.4) and
post-vaccination considerations (Chapter 1.5).

Chapter 1.4 provides detailed sections on the administration of vaccines. This


chapter discusses occupational health and safety issues and the equipment
required for vaccination. Techniques for vaccine administration, including the
route and site of vaccine administration, positioning, identifying the vaccination
site, and methods for administering multiple vaccine injections at the same visit
are described in detail.
Chapter 1.5 provides readers with information on post-vaccination care,
including immediate after-care and the recognition and management of adverse
events following immunisation (AEFI), including anaphylaxis. There are also
sections on how to report AEFI, documentation of vaccination, and details about
the Australian Childhood Immunisation Register.
A summary of the key points discussed in these chapters is provided in the table
in Appendix 10, Summary table procedures for a vaccination encounter. This is
suitable for photocopying for training and auditing purposes.

An overview of vaccination preface to Chapters 1.31.5 7

1.2 An overview of
vaccination preface
to Chapters 1.31.5

Chapter 1.3 describes the steps required in preparing for a vaccination encounter.
This includes preparation of an anaphylaxis response kit and effective cold
chain management (transport, storage and handling of vaccines) as described
in National Vaccine Storage Guidelines: Strive for 5. The next section discusses
obtaining valid consent and is followed by information on comprehensive
pre-vaccination health screening, including a standard screening checklist and
summary tables of precautions and contraindications to vaccination. The chapter
ends with a section on how to manage catch-up vaccination. The section is
divided into two categories; the first for children <8years of age and the second
for people 8years of age.

1.3 Pre-vaccination procedures


The following sections discuss steps and procedures that should occur before a
vaccination encounter. In addition, Appendix 10, Summary table procedures for a
vaccination encounter provides a table summarising the procedures described in
this chapter.

1.3.1 Preparing an anaphylaxis response kit


The availability of protocols, equipment and drugs necessary for the
management of anaphylaxis should be checked before each vaccination session.
An anaphylaxis response kit should be on hand at all times and should contain:
adrenaline 1:1000 (minimum of 3 ampoules check expiry dates),
minimum of three 1mL syringes and 25 mm length needles (for IM injection),
cotton wool swabs,
pen and paper to record time of administration of adrenaline, and
laminated copy of Recognition and treatment of anaphylaxis (back cover of
thisHandbook).
Section 1.5.2 provides details on recognition and treatment of adverse events
following immunisation.

1.3.2 Effective cold chain: transport,


storage and handling of vaccines1,2
The cold chain is the system of transporting and storing vaccines within the
temperature range of +2C to +8C from the place of manufacture to the point of
administration.
All immunisation service providers should be familiar with and adhere to the
National Vaccine Storage Guidelines: Strive for 5. This publication can be accessed
free of charge from http://www.immunise.health.gov.au/internet/immunise/
publishing.nsf/Content/provider-store
The National Vaccine Storage Guidelines: Strive for 5 contain specific details
on setting up the infrastructure for a vaccination service, and immunisation
service providers should refer to this to ensure that satisfactory equipment and
procedures are in place before commencing vaccination services. The Guidelines
also provide instructions on how to best transport vaccines from the main storage
facility to outreach or external clinics using a cooler.
With correct temperature monitoring and adherence to the cold chain Guidelines,
any problems in vaccine storage should be detected early and handled
appropriately before compromised vaccine is administered.

8 The Australian Immunisation Handbook 9th Edition

Purpose-built vaccine refrigerators (PBVR) are the preferred means of storage


for vaccines. Domestic refrigerators are designed and built for food and drink
storage, not for the special temperature needs of vaccines.
Cyclic defrost and bar refrigerators are not recommended because they produce
wide fluctuations in internal temperatures and regular internal heating.
Bar refrigerators, in particular, should not be used because of the risk of freezing,
temperature instability and susceptibility to ambient temperatures.
If the only alternative is to use a domestic refrigerator for vaccine storage,
modification of the refrigerator is essential to reduce the risk of adverse vaccine
storage events. Please refer to the cold chain Guidelines for further information.
The following checklist summarises the ongoing activities required by
immunisation service providers to ensure optimal storage of vaccines:
(a) Ensure one staff member is designated as administrator of vaccines and
vaccine storage; only one staff member should be responsible for refrigerator
thermostat controls at any one time.
(b) Name a back-up vaccine administrator, to take responsibility for vaccines in
the absence of the primary vaccine administrator.
(c) Ensure your healthcare service has a written Vaccine Management policy
and protocol which, as a minimum, should include:
how and when to monitor and record the minimum and maximum
temperatures of the vaccine refrigerator,

how to order and receive vaccines and rotate stock,


how to store the vaccine, diluents and ice/gel packs correctly in the
refrigerator,
how to maintain the refrigerator including a regimen of regular servicing,
what steps to take if the refrigerator temperature goes outside +2C
to +8C, including identification of a cold chain breach, response
procedures, documentation, recording and prevention of recurrences,
how to manage the vaccines during a power failure,
how to pack a portable cooler properly, including correct conditioning of
ice packs/gel packs,
training required for staff handling vaccines.
(d) Storage of all vaccines:
Maintain refrigerator temperature between +2C to +8C, check and record
the current plus minimum/maximum temperatures at least daily or
immediately before vaccines are used.

Pre-vaccination procedures 9

1.3 Pre-vaccination
procedures

how to check the accuracy of the thermometer and/or the data logger,
and how and when to change the thermometer battery,

Twice-daily temperature checks will give a better indication of any


problems in the refrigerators function and temperature fluctuations over
the course of the day.
Keep the door closed as much as possible.
Ensure one person is responsible for adjusting refrigerator controls and
that all staff are appropriately trained to ensure continuous monitoring.
Establish and document protocols for response to cold chain breaches.
A vaccine storage self-audit should be undertaken by the clinic/practice
at least every 12months.
Most vaccines must be protected from freezing. Diluents must also be
protected from freezing, as freezing could cause tiny cracks within the wall
of the diluent container. Protect all vaccines from UV and fluorescent light.
If vaccines have been exposed to temperatures below +2C or above
+8C, follow the practice protocol for response to a breach of the
cold chain. Isolate vaccines and contact the State/Territory health
authority for advice on the National Immunisation Program vaccines
and the manufacturer/supplier for privately purchased vaccines.
Recommendations for the discarding of vaccines may differ between
health authorities and manufacturers. Do not discard any vaccines until
you discuss the necessary actions.
Performmonthly vaccine stocktake, ensure vaccines with the shortest
expiry date are stored at the front of the refrigerator, record and dispose
of vaccines that have passed the expiry date.
Order appropriate levels of stock to ensure the refrigerator is not
overcrowded and that sufficient doses of vaccine are available until the
arrival of the next order. Monthly usage from previousyears can assist
with more accurate ordering.
Ensure all reception staff are familiar with and adhere strictly to the
practice vaccine delivery protocols, including timely unpacking of
vaccines.
Ensure people purchasing vaccines from a pharmacy understand the
need to handle/transport the vaccines correctly.
Minimum/maximum thermometers and/or loggers should be checked for
accuracy (calibrated) annually. Refer to manufacturer for assistance. Change
the battery in digital minimum/maximum thermometers every 12months.
Ensure the refrigerator is placed out of direct sunlight and the
manufacturers instructions for air circulation around the back and sides
are followed.
Ensure the refrigerator is in a secure area accessible to staff only.

10 The Australian Immunisation Handbook 9th Edition

Ensure the power source is marked clearly in a way to prevent the


refrigerator from being accidentally unplugged or turned off.
During a power failure, monitor the temperature of your refrigerator.
If vaccines are at risk, use alternative storage arrangements with
appropriate monitoring.
(e) Using a purpose-built vaccine refrigerator (PBVR):
PBVRs maintain a stable, uniform and controlled cabinet temperature
unaffected by ambient air temperature, and have a defrost cycle that
allows defrosting without rises in cabinet temperature.
PBVRs have a standard alarm and safety feature alert and good
temperature recovery.
Ensure that the PBVR does not constantly display minimum/maximum
and ambient temperatures. Separate minimum/maximum temperatures
must be used to monitor the refrigerator. There are some PBVRs that
require a daily data-logger download to view temperature data.
PBVRs should alarm if temperatures outside +2C to +8C are reached.
Some PBVRs have a back-plate that may vary in temperature during the
defrost cycle. Ensure vaccines are kept 4 cm from the back-plate. Check
with PBVR manufacturer to ensure that vaccines can be stored in the
bottom of the refrigerator.

Do not overstock or crowd the vaccines by overfilling the shelves. Allow


space between stock for air circulation.
If very small amounts of vaccine are stored in a PBVR it is necessary to
add thermal mass (such as bottles of water) to the vacant space to ensure
even temperature is maintained throughout the refrigerator.
If a chart recorder is used, the chart recorder paper must be changed
every 7days and stored in a safe place for auditing purposes.
(f) Using a domestic refrigerator:
Fill the lower drawers and the door with plastic bottles/containers filled
with water.
Get to know the temperatures throughout the refrigerator by monitoring
and recording to identify any cold spots.
Store the vaccines in an enclosed plastic container, in their original
packaging, and label the containers clearly.
Vaccines must never be stored in the door of the refrigerator.

Pre-vaccination procedures 11

1.3 Pre-vaccination
procedures

In the event of a power failure, PBVRs with glass doors will lose their
cool temperature quickly. Ensure the protocol for responding to power
failures is up-to-date and that staff are aware of the procedures.

Ensure each domestic refrigerator has a Celsius digital minimum/


maximum thermometer, with the thermometer probe placed inside
vaccine packaging, inside and near the back of an enclosed plastic
container. A temperature recording chart is also required.

Cold chain breaches


Do not use vaccines exposed to temperatures below +2C or above +8C
without obtaining further advice. Do not discard these vaccines. Isolate
vaccines and contact the State/Territory health authorities for advice on the
National Immunisation Program vaccines and the manufacturer/supplier
for privately purchased vaccines. Recommendations for the discarding of
vaccines may differ between health authorities and manufacturers. Do not
discard any vaccines until you discuss the necessary actions.

1.3.3 Valid consent


Valid consent can be defined as the voluntary agreement by an individual to a
proposed procedure, given after appropriate and reliable information about the
procedure, including the potential risks and benefits, has been conveyed to the
individual.3-7

For consent to be legally valid, the following elements must be present:8


It must be given by a person with legal capacity, and of sufficient intellectual
capacity to understand the implications of being vaccinated.
It must be given voluntarily.
It can only be given after the relevant vaccine(s) and their potential risks and
benefits have been explained to the individual.
The individual must have sufficient opportunity to seek further details or
explanations about the vaccine(s) and/or their administration.
Consent should be obtained before each vaccination, once it has been established
that there are no medical conditions that contraindicate vaccination.

Consent on behalf of a child or adolescent


In general, a parent or legal guardian of a child has the authority to consent to
vaccination of a child.3,7 A child in this context is defined as being under the age
of 18years in all States and Territories except New South Wales, where the age
is 14years, and in South Australia and the Northern Territory, where the age is
16years.
If they are of sufficient age and maturity to understand the proposed procedure and
the risks and benefits associated with same, children at younger ages may be able to
provide consent for procedures such as vaccination. Please refer to your own State
or Territory immunisation service provider guidelines for more information.

12 The Australian Immunisation Handbook 9th Edition

Should a child or adolescent refuse vaccinations for which a parent/guardian has


given consent, the childs wishes should be respected and the parent/guardian
informed.3

Consent on behalf of people with impaired decision-making ability


A responsible adult family member, preferably with authority to make medical
decisions, may give consent for vaccination of an adult with a significant
disability. For example, this may occur for influenza vaccination of an elderly
person with dementia.

Resources to help communicate the risks and benefits of vaccines


Plain language should be used in communicating information about vaccines
and their use to an individual. The individual must be allowed to ask for further
information and have time to make a decision about whether to consent or not.9,10
It is preferable that printed information is available to supplement any verbal
explanations.11 The summary table Comparison of the effects of diseases and the side
effects of vaccines inside the front cover of this Handbook provides some basic
information necessary to communicate the risks and benefits of vaccination. The
table can be photocopied and used freely as required.
More detailed information concerning vaccines and their use is available from the
following sources:

www.ncirs.usyd.edu.au
The National Centre for Immunisation Research and Surveillance of Vaccine
Preventable Diseases website includes fact sheets related to specific vaccines,
vaccine-preventable diseases and vaccine safety.
See also Appendix 5, Commonly asked questions about vaccination.

Evidence of consent
General practice or public immunisation clinics
Consent may be given either in writing or verbally, according to the protocols
of the health facility, but it must meet the criteria for valid consent. Evidence
of verbal consent should be documented in the clinical records. If a standard
procedure is routinely followed in a practice or clinic, then a stamp, a sticker or a
providers signature indicating that the routine procedure has been followed, may
be used. For paperless medical records, a typed record of verbal consent may be
made in the patients file, or a copy of written consent scanned into the file.

Pre-vaccination procedures 13

1.3 Pre-vaccination
procedures

www.immunise.health.gov.au
The Immunise Australia website includes Common questions and answers
(fact sheets), Understanding childhood immunisation and links to State
and Territory Health Department websites. Several of these sites offer
multilingual fact sheets.

Consent is often given and recorded at the first vaccination visit. Explicit verbal
consent is required before subsequent vaccinations even when written consent
has been given at previous vaccination encounters.
School-based vaccination programs
Consent is often given for the entire vaccination program and is valid for the
number of doses to be given during a school-based vaccination program.
In school-based (and other large-scale) vaccination programs, the parent or
guardian usually does not attend with the child on the day the vaccination is
given, and written consent from the parent or guardian is desirable in these
circumstances. However, if further clarification is required, verbal consent may
be sought by telephone from the parent or guardian by the immunisation service
provider. This should be clearly documented on the childs consent form. Older
adolescents may be able to provide their own consent for vaccinations.12 However,
the vaccination program may vary between jurisdictions. Please refer to your own
State or Territory immunisation service provider guidelines for more information.

1.3.4 Pre-vaccination screening


Immunisation service providers should perform a pre-vaccination health screen
of all recipients to determine:
if there are any contraindications or precautions to the vaccines that are to be
administered, and
whether alternative or additional vaccines should be considered.
For some individuals, alterations to the routinely recommended vaccines may be
necessary to either eliminate or minimise the risk of adverse events, to optimise an
individuals immune response, or to enhance the protection of a household contact
against vaccine-preventable diseases.
Such changes to the recommended vaccines may require discussion with an
immunisation expert such as the local immunisation coordinator or a medical
practitioner with expertise in vaccination.

14 The Australian Immunisation Handbook 9th Edition

Steps for pre-vaccination screening


A comprehensive pre-vaccination health screening is necessary to assess a
persons medical fitness for vaccination and to determine whether a different
vaccine schedule may be recommended. Follow these steps to complete the
screening process:
1. Provide the person to be vaccinated or the parent/carer with the Prevaccination screening checklist (Table 1.3.1). NB. Some of the questions in
this checklist are deliberately non-specific so as to elicit as much important
information as possible.
The pre-vaccination screening checklist may be photocopied and handed
to the parent/carer or person to be vaccinated just before vaccination.
It may also be photocopied and displayed in the clinic/surgery for easy
reference for the immunisation service provider.
2. When any of the conditions or circumstances are identified by using the
pre-vaccination screening checklist, refer then to Table 1.3.2 which lists the
specific issues pertaining to these conditions or circumstances and provides
the appropriate action with a rationale.
3. Where necessary, further expert advice should be sought from a medical
practitioner with expertise in vaccination, the immunisation section within
your State or Territory health authority, or your local Public Health Unit (see
Appendix 1, Contact details for Australian, State and Territory Government health
authorities and communicable disease control).

Pre-vaccination procedures 15

1.3 Pre-vaccination
procedures

4. No one should be denied the benefits of vaccination by withholding vaccines


for inappropriate reasons (see Table 1.3.4 False contraindications to vaccination).

Table 1.3.1: Pre-vaccination screening checklist


Pre-vaccination screening checklist
This checklist helps your doctor/nurse decide about vaccinating you or your child.
Please tell your doctor/nurse if the person about to be vaccinated:
is unwell today
has a disease which lowers immunity (eg. leukaemia, cancer, HIV/AIDS)
or is having treatment which lowers immunity (eg. oral steroid medicines
such as cortisone and prednisone, radiotherapy, chemotherapy)
has had a severe reaction following any vaccine
has any severe allergies (to anything)
has had any vaccine in the pastmonth
has had an injection of immunoglobulin, or received any blood
products or a whole blood transfusion within the past year
is pregnant
has a past history of Guillain-Barr syndrome
was a preterm infant
has a chronic illness
has a bleeding disorder
A different vaccine schedule may be recommended if the person to be vaccinated:
identifies as an Aboriginal or Torres Strait Islander
does not have a functioning spleen
is planning a pregnancy or anticipating parenthood
is a parent, grandparent or carer of a newborn
lives with someone who has a disease which lowers immunity (eg. leukaemia, cancer,
HIV/AIDS), or lives with someone who is having treatment which lowers immunity (eg.
oral steroid medicines such as cortisone and prednisone, radiotherapy, chemotherapy)
Note: Please ask your doctor/nurse questions about this information or any
other matter relating to vaccination before the vaccines are given.
Before any vaccination takes place, the immunisation service provider will ask you:
Did you understand the information provided to you about immunisation?
Do you need more information to decide whether to proceed?
Did you bring your/your childs vaccination record card with you?
It is important for you to receive a personal record of your or your childs
injections. If you do not have a record, ask your immunisation service provider
to give you one. Bring this record with you every time you or your child visit
for vaccination. Make sure your doctor/nurse records all vaccinations on it.
Your child may need this record to enter childcare, preschool or school.

16 The Australian Immunisation Handbook 9th Edition

Conditions or circumstances identified using


the pre-vaccination screening checklist
The recommended responses for immunisation service providers to any
conditions or circumstances identified by the pre-screening checklist is
summarised in Table 1.3.2. NB. Only vaccines recommended on the National
Immunisation Program schedule are included in Table 1.3.2. For information on
other vaccines, refer to the relevant chapter within this Handbook (Part3) or to
vaccine product information. For reference, Table 1.3.3 provides a classification of
live attenuated vaccines.
Table 1.3.2: Responses to relevant conditions or circumstances identified by the
pre-vaccination screening checklist
Condition or circumstance

Action

Rationale13-15

Unwell today:

Defer all vaccines


until afebrile.

To avoid an adverse event


in an already unwell child,
or to avoid attributing
symptoms to vaccination.

Acute febrile illness


(current T 38.5C).
Acute systemic illness.

Has a disease which


lowers immunity or
receiving treatment which
lowers immunity.

Seek expert advice


before vaccination
(see Appendix 1).
NB. People living with
someone with lowered
immunity should be
vaccinated, including
with live viral vaccines.

Anaphylaxis following
a previous dose of the
relevant vaccine.

Do not vaccinate.

A severe (anaphylactic)
allergy to a vaccine
component.

Do not vaccinate (seek


specialist advice as
per Appendix 1).

Refer to Appendix 4
for vaccine component
checklist.

See also Contraindications


to vaccination below.

Received live parenteral


vaccine or BCG vaccine
in past 4weeks.

Delay live vaccines


by 4weeks.

See also Contraindications


to vaccination below.

The safety and effectiveness


of the vaccine may be
suboptimal in people with
impaired immunity.

Anaphylaxis to a
previous dose of vaccine
is a contraindication to
receiving the vaccine.
Anaphylaxis to a
vaccine component is
a contraindication to
receiving the vaccine.

The immune response to a


live viral vaccine may interfere
with the response to a second
live viral vaccine if given
within 4weeks of the first.

Pre-vaccination procedures 17

1.3 Pre-vaccination
procedures

See Section 2.3.3,


Vaccination of individuals
with impaired immunity
due to disease or treatment.

NB. Children with minor


illnesses (without acute
systemic symptoms/signs)
should be vaccinated.

Condition or circumstance

Action

Rationale13-15

Has had any blood


product in the past
7months, or has had IM
or IV immunoglobulin
in the past 11months.

Make a return appointment


for this vaccination,
and send a reminder
later if necessary.

Antibodies within these


products may interfere
with the immune response
to these vaccines.

Live vaccines* should be


deferred until after delivery.

There is insufficient evidence


to ensure the safety of
administering live vaccines
during pregnancy or within
28days before conception.

Refer to Table 2.3.5


Recommended
intervals between either
immunoglobulins or blood
products and MMR, MMRV
or varicella vaccination.
Is pregnant.
Refer to Table 2.3.1
Vaccinations in pregnancy.

History of Guillain-Barr
syndrome (GBS).
See Chapter 3.9, Influenza.

Was born preterm.


See Section 2.3.2,
Vaccination of women
planning pregnancy,
pregnant or breastfeeding
women, and preterm infants.

Conception should
be deferred until at
least 28days after
administration of live
viral vaccines. Inactivated
vaccines are generally
not contraindicated
in pregnancy.

NB. Influenza vaccine


is recommended for
pregnant women.
Vaccination of household
contacts of pregnant
women should be
completed according to
the NIP schedule.

Risks and benefits of


influenza vaccine should
be weighed against the
potential risk of GBS
recurrence (seek specialist
advice as per Appendix 1).

People with a history of


GBS may be at risk of
recurrence of the condition
following influenza vaccine.

Preterm infants born at


<28weeks gestation or
<1500g birth weight
require an extra dose of
PRP-OMP Hib vaccine
at 6months of age.

Preterm infants may be at


increased risk of vaccinepreventable diseases (eg.
invasive pneumococcal
disease (IPD)), and may not
mount an optimal immune
response to certain vaccines
(eg. hepatitis B, PRP-OMP).

Preterm infants born


at <28weeks gestation
and/or with chronic lung
disease require extra
pneumococcal vaccinations.
Preterm infants born at
<32weeks gestation or
<2000g birth weight may
require an extra dose of
hepatitis B vaccine.

18 The Australian Immunisation Handbook 9th Edition

Condition or circumstance

Action

Rationale13-15

Has a severe or
chronic illness.

These people should


receive pneumococcal
vaccine and annual
influenza vaccination.

People with a severe or


chronic illness may be at
increased risk of vaccinepreventable diseases (eg.
IPD) but may not mount an
optimal immune response
to certain vaccines.

See Chapter 2.3, Groups


with special vaccination
requirements.

Has a bleeding disorder.


See Section 2.3.6,
Vaccination of patients
with bleeding disorders.

Identifies as an Aboriginal
or Torres Strait Islander.
See Chapter 2.1, Vaccination
for Aboriginal and Torres
Strait Islander people.
Does not have a
functioning spleen.

Is planning a pregnancy or
anticipating parenthood.

Is a parent, grandparent
or carer of a newborn.

The subcutaneous route


could be considered as
an alternative to the
intramuscular route
(seek specialist advice
as per Appendix 1).

Intramuscular injection
may lead to haematomas
in patients with disorders
of haemostasis.

See the National


Immunisation Program
Indigenous schedules.

Some groups of Indigenous


people are at increased risk
of some of the vaccinepreventable diseases.

Check vaccination status


for pneumococcal,
meningococcal and
Hib vaccinations.

Individuals with an absent


or dysfunctional spleen
are at an increased risk of
severe bacterial infections,
most notably IPD.

Ensure prospective parents


have been offered vaccines
recommended for their agegroup including 2nd dose of
MMR if born after 1966, and
dTpa (unless they have had
a previous dose of dTpa).

Vaccinating before pregnancy


may prevent maternal illness
which could affect the infant,
and may confer passive
immunity to the newborn.

Ensure parents,
grandparents and carers
of a newborn have been
offered all vaccines
recommended for their
age-group including dTpa
(unless they have had a
previous dose of dTpa).

People in close contact are


the most likely sources
of vaccine-preventable
diseases, in particular
pertussis, in the newborn.

NB. Advise women not to


become pregnant within
28days of receiving
live viral vaccines.

Pre-vaccination procedures 19

1.3 Pre-vaccination
procedures

See Section 2.3.3,


Subsection 2.3.3.5,
Individuals with functional
or anatomical asplenia.

If there is significantly
impaired immunity,
they should not receive
live vaccines, but
inactivated vaccines
should be considered
(seek expert advice).

Condition or circumstance

Action

Rationale13-15

Lives with someone who


has impaired immunity.

Ensure all vaccines (in


particular MMR, varicella
and influenza vaccines)
recommended for their
age-group have been
offered to household
members of people with
impaired immunity.

Household members are


the most likely sources
of vaccine-preventable
diseases among people
with impaired immunity
(who often are unable to
be vaccinated, especially
with live viral vaccines).

* Live attenuated vaccines are classified in Table 1.3.3 below.


See Chapter 3.3, Diphtheria, Chapter 3.14, Pertussis or Chapter 3.21, Tetanus for further
information.

Table 1.3.3: Live attenuated parenteral and oral vaccines


Live attenuated parenteral vaccines

Live attenuated oral vaccines

Viral

Bacterial

Viral

Bacterial

MMR

BCG

Oral rotavirus vaccine

Oral typhoid vaccine

MMRV
Varicella vaccine (VV)
Monovalent
rubella vaccine
Yellow fever

Contraindications to vaccination
There are only 2 absolute contraindications applicable to all vaccines:
(i) anaphylaxis following a previous dose of the relevant vaccine, and
(ii) anaphylaxis following any component of the relevant vaccine.
There are 2 further contraindications applicable to live (both parenteral and
oral) vaccines:
(iii) Live vaccines should not be administered to individuals with impaired
immunity, regardless of whether the impairment is caused by disease
or treatment. The exception is that, with specialist advice, MMR can be
administered to HIV-infected individuals in whom impaired immunity
is mild. (See Section 2.3.3, Vaccination of individuals with impaired immunity
due to disease or treatment, and individual vaccine chapters.)
(iv) In general, live vaccines should not be administered during pregnancy,
and women should be advised not to become pregnant within 4weeks of
receiving a live vaccine (see Table 2.3.1 Vaccinations in pregnancy).

20 The Australian Immunisation Handbook 9th Edition

False contraindications to vaccination


Conditions listed in Table 1.3.4 below are not contraindications to vaccination.
People with these conditions should be vaccinated with all recommended
vaccines.
Table 1.3.4: False contraindications to vaccination
The following conditions are not contraindications to any of the
vaccines in the National Immunisation Program schedule:
mild illness without fever (T <38.5C),
family history of any adverse events following immunisation,
past history of convulsions,
treatment with antibiotics,
treatment with locally acting (inhaled or low-dose topical) steroids,
replacement corticosteroids,
asthma, eczema, atopy, hay fever or snuffles,
previous pertussis-like illness, measles, rubella, mumps or meningococcal disease,
prematurity (vaccination should not be postponed),
history of neonatal jaundice,
low weight in an otherwise healthy child,
any neurological conditions including cerebral palsy and Down syndrome,
contact with an infectious disease,
childs mother is pregnant,
woman to be vaccinated is breastfeeding,
recent or imminent surgery,
poorly documented vaccination history.

1.3.5 Catch-up
Every opportunity should be taken to review an individuals vaccination history
and, based on documentation, administer the appropriate vaccine(s). If the
individual has not received vaccines scheduled in the National Immunisation
Program appropriate for his/her age, plan and document a catch-up schedule and
discuss this with the individual. The assessment of vaccination status should be
based on the schedule for the State/Territory in which the individual is residing.
The objective of catch-up vaccination is to complete a course of vaccination and
provide optimal protection as quickly as possible. The information and tables
below will assist in planning a catch-up schedule. If the immunisation service
provider is still uncertain about how to plan the catch-up schedule, expert advice
should be sought (see Appendix 1, Contact details for Australian, State and Territory
Government health authorities and communicable disease control).

Pre-vaccination procedures 21

1.3 Pre-vaccination
procedures

child to be vaccinated is being breastfed,

An on-line catch-up calculator is available at


www.health.sa.gov.au/immunisationcalculator
This calculator is regularly updated for all catch-up scenarios relevant to the
NIP. For non-NIP vaccines or complicated catch-up scenarios, expert advice
should be sought (see Appendix 1, Contact details for Australian, State and
Territory Government health authorities and communicable disease control).
To calculate a catch-up schedule, the on-line calculator requires the childs date
of birth, State of residence, past vaccination history and Indigenous status.
The calculator can be used for children 7years of age (the age up to which
vaccinations will be recorded on the ACIR) and can calculate catch-up schedules
for children from all States and Territories. For recently arrived immigrants,
the World Health Organization web site www.who.int/countries/en lists an
immunisation schedule (where provided by that particular country) and may
supplement information regarding which vaccines a child/adult may have
received (see also Section 2.3.9, Vaccination of immigrants to Australia).
Alternatively, the instructions and guidelines below will assist in the manual
calculation of a catch-up schedule.

Determining a vaccination history


Individuals with incomplete vaccination records
The most important requirement for assessment of vaccination status is to have
written documentation of vaccination. The approach of providers to the problem
of inadequate records should be based on the age of the individual, whether
previous vaccines have been given in Australia or overseas, and the vaccines
being considered for catch-up.

Vaccines given from 1 January 1996


The Australian Childhood Immunisation Register (ACIR) commenced on 1
January 1996 and all vaccinations given to children since then should be available
from the ACIR. If the parent states that vaccines not recorded on the ACIR have
been given, every effort should be made to contact the relevant immunisation
service provider. If confirmation from the nominated provider or the ACIR
cannot be obtained, and no written records are available, the vaccines should be
considered as not received, and the child should be offered a catch-up course of
vaccination appropriate for age (see Section 1.3.5). Parents can obtain an ACIR
Immunisation History Statement from Medicare (see Section 1.5.4).

Older children and adolescents <18years of age


No vaccination information is recorded on the ACIR after a child turns 7years
of age, but any information already held is retained. The information will relate
only to vaccines received between birth and the 7th birthday. The ACIR Enquiry
Line can be contacted on 1800 653 809 and any record held for an individual who

22 The Australian Immunisation Handbook 9th Edition

is 7years of age can be made available to an immunisation service provider or


parent/carer.
In older children and adolescents, alternative sources of documentation (such
as personal health records) will be needed, but are less likely to be available
with increasing age. Individuals who do not have personal vaccination records
may seek evidence of past vaccination from their parents, their past and present
healthcare providers or immunisation service providers, including Local
Government immunisation service providers. Those born after 1990 may have
some vaccinations recorded on the ACIR (see Section 1.5.4).
For most vaccines, there are no adverse events associated with additional doses
in immune individuals. In the case of diphtheria and tetanus vaccines, additional
doses may occasionally be associated with an increase in local adverse events
in immune individuals (see Chapter 3.3, Diphtheria, Chapter 3.14, Pertussis or
Chapter 3.21, Tetanus). However, the benefits of protection against pertussis are
likely to outweigh the risk of an adverse reaction.

Adults (18years of age)

Guidelines for planning catch-up vaccination


There are a number of tables in this section which are designed to help plan a
catch-up schedule if not using the on-line calculator.
Figure 1.3.1 is a worksheet for calculating and recording which vaccines are
required, the number of doses outstanding and the timing of these doses.
Table 1.3.5 can be used to assess the number of doses a child would have
received if they were on schedule. Check under the current age of the child
to see how many doses they should have already received and use that
number of doses as the starting point for calculating a catch-up schedule. For
example, a child who is 18months old now should have received 3 doses of
DTPa, 3 doses of IPV etc.
Table 1.3.6 lists the minimum interval between doses.
Tables 1.3.81.3.11 are for calculating catch-up for Hib and pneumococcal
vaccination.
If documentation cannot be produced, assume that the vaccine has not been given
previously, unless contact can be made with the immunisation service provider.

Pre-vaccination procedures 23

1.3 Pre-vaccination
procedures

In adults, written documentation of previous vaccination history may not be


available. It is important, however, to seek information of any previous doses
of diphtheria and tetanus vaccines, and of pneumococcal polysaccharide
vaccination in the previous 5years, as increased local reactions may occasionally
occur in immune individuals (see Chapter 3.3, Diphtheria, Chapter 3.15,
Pneumococcal disease or Chapter 3.21, Tetanus). Additional doses of MMR,
varicella, IPV or hepatitis B vaccine are rarely associated with significant adverse
events in adults.

Vaccine doses should not be administered at less than the recommended


minimum interval16 (see Table 1.3.6).
In exceptional circumstances, where early vaccination is required, Table 1.3.7
indicates the minimum age that the first dose of a vaccine may be given.
Doses administered earlier than the minimum interval should not be
considered as valid doses and should be repeated as appropriate using
Table1.3.5.
When commencing the catch-up schedule, the standard scheduled interval
between doses may be reduced or extended, and the numbers of doses
required may reduce with age. For example, from 15months of age, only 1
dose of (any) Hib vaccine is required.
As a child gets older, the recommended number of vaccine doses may change
(or even be omitted from the schedule), as the child becomes less vulnerable
to specific diseases.
For incomplete or overdue vaccinations, build on the previous documented
doses.
Never start the schedule again, regardless of the interval since the last dose.
If more than 1 vaccine is overdue, 1 dose of each due or overdue vaccine
should be given now. Further required doses should be scheduled after the
appropriate minimum interval (see Table1.3.6).
A catch-up schedule may require multiple vaccinations at a visit. Give all the
due vaccines at the same visit do not defer. See Section 1.4.9 for procedures
for administering multiple injections at the same visit.
The standard intervals and ages recommended in the NIP schedule should be
used once the child or adult is up-to-date with the schedule.
Some individuals will require further doses of antigens that are available only
in combination vaccines. In general, the use of the combination vaccine(s)
is acceptable, even if this means the number of doses of another antigen
administered exceeds the required number.
If different Hib vaccines are inadvertently used in the primary series, then
3 doses (of any Hib vaccine) are required at 2, 4 and 6months of age, with a
booster at 12months of age (see Chapter 3.4, Haemophilus influenzae type b).
NB. Routine rotavirus vaccine catch-up of older children is not recommended.
Infants should commence the course of rotavirus vaccination within the
recommended age limits for the first dose. It is also necessary to ensure that
doses are not given beyond the upper age limits for the final dose of the vaccine
course (see Chapter 3.18, Rotavirus).

24 The Australian Immunisation Handbook 9th Edition

Interruption to a vaccination
If the process of administration of a vaccine given parenterally (IM or SC) is
interrupted (eg. by syringe-needle disconnection) the whole dose should be
repeated as soon as practicable.
If an infant regurgitates or vomits part of a dose of oral rotavirus vaccine,
it is not necessary to repeat the dose. Therefore, the regurgitated (and
incomplete volume) dose is still considered as the valid dose (see Chapter
3.18, Rotavirus).

Determining a catch-up schedule for children <8years of age


A catch-up schedule for a child <8years of age should be planned by taking into
account the guidelines above and using Table 1.3.5. The Catch-up Worksheet
(Figure 1.3.1) provides a method of recording these steps. All catch-up vaccines
administered to children aged <7years should be reported as soon as is
practicable to the ACIR.

Using the Catch-up Worksheet


1. Record the childs details including date of birth and current age in the top
left corner of the worksheet.
2. For each vaccine, determine how many doses have been received and the
date that the last dose was given. Record this on the worksheet.

4. Assess other factors that may affect the type or number of vaccines required,
including:
anaphylaxis to any vaccine or one of its components (that vaccine is
contraindicated),
impaired immunity due to disease or treatment (see Chapter 2.3, Groups
with special vaccination requirements),
identifying as an Aboriginal or Torres Strait Islander (see Chapter 2.1,
Vaccination for Aboriginal and Torres Strait Islander people),
children with an underlying medical risk condition which predisposes
them to invasive pneumococcal disease (see Chapter 3.15, Pneumococcal
disease),
a reliable history of previous varicella infection (varicella vaccine not
required), and
babies born at <32weeks gestation (see Hib vaccine and hepatitis B vaccine
catch-up below).

Record any relevant factor in the comments column beside the relevant
vaccine.

Pre-vaccination procedures 25

1.3 Pre-vaccination
procedures

3. Refer to Table 1.3.5 to check how many doses of each vaccine are required for
the childs current age. Enter this number in the appropriate column of the
worksheet.

5. If any variations to the schedule are necessary due to recorded factors (eg.
a child with impaired immunity may require different vaccines), adjust the
number of doses required accordingly.
6. For each vaccine, compare the last dose given with the number required for
the childs current age.
7. If the child has already received the number of doses required, the relevant
dose number due now and further doses cells should be crossed through.
8. If the number of the last dose given is less than the number required, a dose
of the relevant vaccine should be administered now, and recorded in the
dose number due now cell. If this dose still does not complete the required
doses, enter the further dose numbers in the further doses cell.
9. Refer to Table 1.3.6 to determine the recommended minimum intervals
required between doses and record in the relevant further doses cells.
10. Convert this information into a list of proposed appointment dates, detailing
vaccines and dose number needed at each visit on the Catch-up Worksheet.
11. Record this catch-up schedule in your provider records and provide a copy to
the parent/carer.

26 The Australian Immunisation Handbook 9th Edition

Figure 1.3.1: Catch-up Worksheet for children <8years of age


CATCH-UP WORKSHEET
Name:
DOB:

Last dose
given
Dose
number
and date

Number
of doses
required
at current
age*

Dose
number
due now

Further
doses

Comments

Interval
or date

Age:
DTPa
Poliomyelitis
(IPV)
Hepatitis A
Hepatitis B
Hib
7vPCV &
23vPPV
MenCCV
MMR
Rotavirus

DO NOT give after


upper age limits
for each dose.

Varicella
CATCH-UP APPOINTMENTS
Date

Vaccines
& Dose
number

Interval to next dose

Comments

* See step 5 Using the Catch-up Worksheet above.

Pre-vaccination procedures 27

1.3 Pre-vaccination
procedures

See Table 3.18.1.

Table 1.3.5: Number of vaccine doses that should have been administered by
the current age of the child (table to be used in conjunction with Catch-up
Worksheet)
VACCINE

CURRENT AGE
0<2mo

2<4mo

4<6mo

6<12mo

1218mo

>18mo<4yr

4yr<8yr

DTPa*

Poliomyelitis
(IPV)

Hepatitis A
Hepatitis B

birth
dose
given

birth
dose not
given

3^

Hib

Complex see Table 1.3.8 for Hib vaccine catch-up

7vPCV &
23vPPV

Complex see Tables 1.3.9, 1.3.10 and 1.3.11


for pneumococcal vaccine catch-up

MenCCV
MMR
Rotavirus#

There are specific age


limits as per Chapter 3.18,
Rotavirus, Table 3.18.1.

NO CATCH-UP

Varicella

* Some children may have received 4 doses of DTP by 18months of age, especially if moved
from overseas. These children will require a 5th dose of DTPa at 4years of age.
If the 3rd dose of IPV is given after 4years of age, a 4th dose is not required. However, if
using a combination vaccine it is acceptable to receive a 4th dose.
Indigenous children resident in NT, QLD, SA and WA only. Dependent on jurisdiction,
the 1st dose is given at 1218months of age followed by the 2nd dose 6months later at
1824months of age. Consult relevant State/Territory authorities for advice regarding
catch-up in children older than 2years of age.
Birth dose should be given within 7days of birth. Although a birth dose of hepatitis B
vaccine is recommended for all infants, a catch-up dose is not necessary if it was not given.
Even if the birth dose was given, a further 3 doses of hepatitis B vaccine are required.
^ Some States/Territories schedule a 3rd dose (or the 4th dose) of hepatitis B vaccine at
6months of age rather than 12months.
# There is no catch-up for rotavirus vaccine (see Chapter 3.18, Rotavirus).

28 The Australian Immunisation Handbook 9th Edition

Table 1.3.6: Minimum dose intervals for NIP vaccines for children <8years of
age (table to be used in conjunction with Catch-up Worksheet)
Vaccine

Minimum
interval
between dose
1&2

Minimum
interval
between dose
2&3

Minimum
interval
between dose
3&4

DTPa*

4weeks

4weeks

6months

Poliomyelitis (IPV)

4weeks

4weeks

4weeks

Hepatitis A

6months

8weeks

(Indigenous children in NT,


QLD, SA & WA only)
Hepatitis B
If first dose given at birth
or at 7days after birth

4weeks

8weeks

If first dose is not given at birth


or at >7days after birth

4weeks

8weeks

Hib (PRP-OMP)

See Table 1.3.8 Hib vaccine catch-up

Hib (PRP-T)
Pneumococcal (7vPCV)

See Tables 1.3.9, 1.3.10, 1.3.11


Pneumococcal vaccine catch-up

MenCCV^

Rotavirus**

Varicella

4weeks
Rotarix

4weeks

RotaTeq

4weeks

4weeks

4weeks

* If DTPa is only available in combination with other antigens (eg. DTPa-IPV, DTPa-hepBIPV-Hib or DTPa-HepB-IPV), these formulations can be used where necessary for primary
course or catch-up doses in children <8years of age.
If the 3rd dose of IPV is given after 4years of age, a 4th dose is not required. However, if
using a combination vaccine, it is acceptable to receive a 4th dose.
If dose given at birth or within 7days of birth (considered dose 1 for this table), then 3
subsequent doses should be given.
If dose 1 is not given at birth or within 7days of birth, then it should be given at 2months
of age, followed by a further 2 doses.
^ The schedule is a single dose given at 12months of age. Alternative schedules are available
for children <12months of age (see Chapter 3.12, Meningococcal disease).
# MMR vaccine may be given from 9months of age if in contact with case, but dose must be
repeated at 12months of age.
** Consult Chapter 3.18, Rotavirus, Table 3.18.1 for upper age limits for administration of
rotavirus vaccines. Catch-up is not recommended.

Pre-vaccination procedures 29

1.3 Pre-vaccination
procedures

MMR#

Table 1.3.7: Minimum age for the first dose of vaccine in exceptional
circumstances*
Vaccine

Minimum age for first


dose in exceptional
circumstances

Minimum age accepted as


valid by ACIR

DTPa

6weeks

6weeks

Poliomyelitis (IPV)

6weeks

6weeks

Hepatitis A

12months

12months

(Indigenous children in
NT, QLD, SA & WA only)
Hepatitis B

6weeks

6weeks

Hib (PRP-OMP)

6weeks

6weeks
6weeks

Hib (PRP-T)

6weeks

MenCCV

6weeks

MMR

9months

12months

11months

Pneumococcal (7vPCV)

6weeks

6weeks

Rotavirus

6weeks

not stated

Varicella

9months (Varilrix)

not stated

12months^ (Varivax)
* Exceptional circumstances may include infants/children being vaccinated before overseas
travel, or opportunistic vaccination following early attendance to a provider. These ages
may differ from routinely recommended ages of administration under the NIP.
If 2 doses of MenCCV are given before 12months of age, then a booster dose should be
given at 12months of age (see Chapter 3.12, Meningococcal disease).
MMR vaccine may be given from 9months of age if in contact with case, but dose must be
repeated at 12months of age.
If a child receives varicella vaccine at <12months of age, a further dose should be given at
18months of age.
^ Receipt of at least 1 dose of varicella vaccine is recommended from 12months of age.

30 The Australian Immunisation Handbook 9th Edition

Catch-up guidelines for individual vaccines


DTPa
Monovalent pertussis vaccine is not available in Australia. If a child has
received previous doses of DT and requires pertussis catch-up, then DTPa or
DTPa-combination vaccines can be used provided that no more than 6 doses of
diphtheria and tetanus toxoids are given before the 8th birthday.
NB. If no birth dose of hepatitis B vaccine was given, and a DTPa-hepatitis
B-containing combination vaccine is used, there should be a minimum interval of
8weeks between doses 2 and 3.
Hepatitis B vaccine
If the infant received the birth dose of hepatitis B vaccine, catch-up doses can be
given 48weeks apart.
If the infant did not receive the birth dose, a catch-up of this dose is not
necessary. In this circumstance, hepatitis B vaccination should commence at
2months of age. There should be a minimum interval of 8weeks between doses 2
and 3.
In preterm babies under 32weeks gestation at birth or <2000g birth weight, it
is recommended to give hepatitis B vaccine at 0, 2, 4 and 6months of age, and
either:
(a) measure anti-HBs at 7months of age and give a booster at 12months of age if
antibody titre is <10 mIU/mL, or
(See also Section 2.3.2, Vaccination of women planning pregnancy, pregnant or
breastfeeding women, and preterm infants and Chapter 3.6, Hepatitis B).
Hib vaccine
The recommended number of doses and recommended intervals of Hib vaccines
vary with the vaccine type and with the age of the child (see Table 1.3.8). PRPOMP is the Hib formulation contained in Liquid PedvaxHIB and COMVAX.
PRP-T is the Hib formulation contained in Hiberix and Infanrix hexa.
Where possible, the same brand of Hib vaccine should be used for all doses. If
different Hib vaccines are used in the primary series, then 3 doses (of any Hib
vaccine) are required at 2, 4 and 6months of age, with a booster at 12months of
age. Only 1 dose (of any Hib vaccine) is required after 15months of age.
When PRP-OMP is used in an extremely preterm baby (<28weeks gestation
or <1500g birth weight), an additional dose should be given at 6months of
age, ie. doses should be given at 2, 4, 6 and 12months of age (see Section 2.3.2,
Vaccination of women planning pregnancy, pregnant or breastfeeding women, and
preterm infants).

Pre-vaccination procedures 31

1.3 Pre-vaccination
procedures

(b) give a booster at 12months of age without measuring the antibody titre.

MMR vaccine
If no previous documented doses have been given, catch-up for MMR consists of
2 doses given at least 4weeks apart.
MenCCV
MenCCV is recommended on the NIP for children at 12months of age. If no
dose was received at 12months of age or if all doses have been received at
<12months of age, a single dose of any meningococcal conjugate vaccine is
recommended (see Chapter 3.12, Meningococcal disease).
7vPCV
The number of doses and recommended intervals of 7vPCV for catch-up vary
with the age of the child, health and Indigenous status of the child, as well as the
State/Territory of residence (see Tables 1.3.9, 1.3.10 and 1.3.11 below).
Low-risk children (including all Indigenous children) aged 2years of age do not
require catch-up.
If <2years of age at presentation, use Table 1.3.9 for low-risk children (including
Indigenous children living in the Australian Capital Territory, New South Wales,
Victoria and Tasmania) and Table 1.3.10 for Indigenous children residing in the
Northern Territory, Queensland, South Australia and Western Australia. Table
1.3.11 provides catch-up details for children aged 5years with an underlying
medical condition. Please also refer to Chapter 3.15, Pneumococcal disease for
further details.
Poliomyelitis vaccine
If no previous documented doses of poliomyelitis vaccine have been given, give
3 doses of IPV or IPV-containing vaccines at least 4weeks apart. (Previous doses
of OPV are interchangeable with IPV.)
If the third dose of IPV is administered before 4years of age, give the fourth
(booster) dose at either the 4th birthday or 4weeks after the third dose, whichever
is later. If the third dose is given after the 4th birthday, a fourth dose is not
required. However, if the use of combination vaccines is necessary, a further IPVcontaining dose may be given.
Rotavirus vaccine
Infants should commence the course of rotavirus vaccination within the
recommended age limits for the first dose, that is by either 12 or 14weeks of age
depending on the vaccine to be used. It is recommended that vaccine doses are
not given beyond the upper age limits specified in Table 3.18.1, Chapter 3.18,
Rotavirus.
Varicella vaccine
If a child receives varicella vaccine at <12months of age, a further dose should be
given at 18months of age.

32 The Australian Immunisation Handbook 9th Edition

Table 1.3.8: Recommendations for Hib catch-up vaccination for children


<5years of age when doses have been delayed or missed
Previous
vaccination
history

Age at
presentation

Type
of Hib
vaccine to
be used

1st dose

2nd dose

3rd dose

Booster dose

0 doses

36 months

PRP-OMP

Give now

1 month
later

Not needed

12 months of age

PRP-T

Give now

1 month
later

12 months
later

12 months of age

PRP-OMP

Give now

2 months
later

Not needed

12 months of
age or 2 months
after 2nd dose
(whichever is
later)

PRP-T

Give now

2 months
later

Not needed

12 months of
age or 2 months
after 2nd dose
(whichever is
later)

711 months

1214 months

Give now

Not needed

Not needed

2 months later

Give now

Not needed

Not needed

18 months of age

1559 months

PRP-OMP
or PRP-T

Give now

Not needed

Not needed

Not needed

36 months

PRP-OMP

PRP-OMP
previously
given

Give now

Not needed

12 months of age

PRP-T

Either
PRP-OMP
or PRP-T
previously
given

Give now

12 months
later

12 months of age

714 months

PRP-OMP
or PRP-T

Previously
given

Give now

Not needed

12 months of
age or 2 months
after 2nd dose
(whichever is
later)

1559 months

PRP-OMP
or PRP-T

Previously
given

Not needed

Not needed

Give now*

2 previous
doses of PRPOMP

1259 months

PRP-OMP
or PRP-T

Previously
given

Previously
given

Not needed

At least 2
months after last
dose*

2 previous
doses of
PRP-T (or
1 of each of
PRP-OMP
and PRP-T)

714 months

PRP-OMP
or PRP-T

Previously
given

Previously
given

At least 1
month after
last dose

1218 months
of age, at least
2months after
last dose

1559 months

PRP-OMP
or PRP-T

Previously
given

Previously
given

Not needed

At least 2
months after last
dose*

1 previous
dose (given at
least 4 weeks
previously)

*A booster dose is not needed if the last previous dose was given at >15months of age.

Pre-vaccination procedures 33

1.3 Pre-vaccination
procedures

PRP-OMP
PRP-T

Table 1.3.9: Recommendations for pneumococcal catch-up vaccination for lowrisk children (including Indigenous children living in ACT, NSW, VIC and
TAS) <2years of age, when doses have been delayed or missed
CATEGORY

Previous doses
of 7vPCV

Age at
presentation

1st dose
7vPCV

2nd dose
7vPCV

3rd dose
7vPCV

All nonIndigenous
children

None

36months

Give now

1month later

12months
later*

717months

Give now

12months
later*

Not needed

and
Indigenous
children living in
ACT, NSW, VIC
and TAS

1823months

Give now

Not needed

Not needed

1 previous
dose (given at
least 4weeks
previously)

511months

Previously
given

Give now

12months
later*

1223months

Previously
given

Give now

Not needed

2 doses

711months

Previously
given

Previously
given

Give now

1223months

Previously
given

Previously
given

Not needed

* Catch-up doses of 7vPCV can be given a minimum of 1month apart to infants aged
<12months. For children aged 12months, there should be a 2month interval between
doses of 7vPCV.

34 The Australian Immunisation Handbook 9th Edition

Table 1.3.10: Recommendations for pneumococcal catch-up vaccination for


Indigenous children <2years of age in NT, QLD, SA and WA, when doses have
been delayed or missed
CATEGORY

Previous
doses of
7vPCV

1st dose
7vPCV

2nd dose
7vPCV

3rd dose
7vPCV

23vPPV*

36 months

Give now

1 month
later

12
months
later

1824 months of age

717 months

Give now

12
months
later

Not
needed

1824 months of age


or 2 months after
2nd dose of 7vPCV
(whichever is later)

1823 months Give now

Not
needed

Not
needed

1824 months of age


or 2 months after
1st dose of 7vPCV
(whichever is later)

1 dose
511 months Previously
(given
given
at least
4 weeks
previously) 1223 months Previously
given

Give now

12
months
later

1824 months of age

Give now

Not
needed

1824 months of age


or 2 months after
2nd dose of 7vPCV
(whichever is later)

2 doses

Previously
given

Previously
given

Give now

1824 months of age

1223 months Previously


given

Previously
given

Not
needed

1824 months of age


or 2 months after
2nd dose of 7vPCV
(whichever is later)

Indigenous
None
children living
in NT, QLD,
SA and WA

711 months

* The timing of 23vPPV varies between States and Territories. Contact your State or Territory
health authority for the appropriate timing.
Catch-up doses of 7vPCV can be given a minimum of 1month apart to infants aged
<12months. For children aged 12months, there should be a 2month interval between
doses of 7vPCV.

Pre-vaccination procedures 35

1.3 Pre-vaccination
procedures

Age at
presentation

36 The Australian Immunisation Handbook 9th Edition

3 doses

2 doses

1259 months

Previously
given

Previously
given

1259 months

Previously
given

1259 months
Previously
given

Previously
given

711 months

711 months

Previously
given

Give now

1259 months

56 months

Give now

711 months

1 dose

Give now

36 months

None

1st dose
7vPCV

Age at
presentation

Previous
doses of
7vPCV

Previously
given

Previously
given

Previously
given

Give now

Give now

Give now

2 months
later

12
months
later

1 month
later

2nd dose
7vPCV

Previously
given

Give now

Give now

Not
needed

Not
needed

1 month
later

Not
needed

Not
needed

12
months
later

3rd dose
7vPCV

Give now

Not needed

12 months of age or 2months after 3rd


dose of 7vPCV (whichever is later)

Not needed

12 months of age or 2months after 2nd


dose of 7vPCV (whichever is later)

12 months of age

Not needed

45 years of age or 2 months after booster


dose of 7vPCV (whichever is later)

45 years of age or 2 months after 3rd dose


of 7vPCV (whichever is later)

45 years of age

45 years of age or 2 months after 2nd


dose of 7vPCV (whichever is later)

45 years of age

45 years of age

45 years of age or 2 months after 2nd


dose of 7vPCV (whichever is later)

45 years of age

45 years of age

12 months of age

12 months of age or 2months after 2nd


dose of 7vPCV (whichever is later)

23vPPV

Booster dose 7vPCV

Catch-up doses of 7vPCV can be given a minimum of 1month apart to infants aged <12months. For children aged 12months, there should be
a 2month interval between doses of 7vPCV.

* Children up to the age of 10years who, after the 6th birthday, develop asplenia, HIV infection, or a haematological malignancy, or who receive a transplant,
should receive 2 doses of 7vPCV 2months apart, and a dose of 23vPPV 2months later. If these children need catch-up doses of 7vPCV, the recommendations
are the same as for the 1259month age-group in Table 1.3.11, with a dose of 23vPPV 2months after the last dose of 7vPCV. See Chapter 3.15, Pneumococcal
disease recommendations and Table 3.15.1

Children 5
years of age
with underlying
medical
conditions

CATEGORY

Table 1.3.11: Recommendations for pneumococcal catch-up vaccination for children 5years of age* with underlying medical conditions

Catch-up schedules for children 8years


of age, adolescents and adults
Catch-up is much less commonly required for these age groups than for young
children. Nevertheless, issues surrounding booster doses or revaccinations are
common, particularly in adults. People who escaped natural infection as children
and were not vaccinated remain at unnecessary risk of vaccine-preventable
diseases.
If a vaccine course is incomplete, never start the course again, regardless of the
interval since the last dose.
Recommendations on vaccination for adults at occupational risk or in a
special risk group can be found in Chapter 2.3, Groups with special vaccination
requirements.
Use Table 1.3.12 to determine:
how many doses of a particular vaccine a person should have received to be
considered completely vaccinated (column 2: Doses required),
deduct any previous doses of the vaccine from that number, and
go to the appropriate minimum interval column.
For example, a 32-year-old woman who has received only 1 dose of hepatitis B
vaccine, 4 doses of the oral poliomyelitis vaccine, 1 dose of MMR vaccine and 2
doses of DTPw as a child, would require:
2 adult doses of hepatitis B, 1 dose given now and a further dose in 8weeks,
no further doses of poliomyelitis vaccine (is fully vaccinated against
poliomyelitis),
varicella vaccine if non-immune,
1 dose of MMR vaccine.
Where several vaccines are required, eg. dTpa, hepatitis B and poliomyelitis
vaccines, never use the available childhood combination vaccines as the antigen
content differs and may result in a severe adverse event. The childhood
combination vaccines are not registered for use in children aged 8years,
adolescents or adults.

Pre-vaccination procedures 37

1.3 Pre-vaccination
procedures

1 dose of dT (preferably given as dTpa),

Table 1.3.12: Catch-up schedules for individuals 8years of age


Vaccine

dT (dTpa*)

Doses required

Minimum interval Minimum interval


between Dose 1
between Dose 2
&2
&3

3 doses

4weeks

4weeks

Hepatitis B

Aged
819years

3 paediatric doses

4weeks

8weeks

Hepatitis B

Aged
1115years
only

2 adult doses

46months

Not required

Hepatitis B

Aged
20years

3 adult doses

4weeks

8weeks

IPV

3 doses

4weeks

4weeks

Human papillomavirus
(females aged
1026years only)

3 doses

4weeks

3months

MMR

2 doses

4weeks

Not required

Varicella vaccine

At least 1 dose if
aged <14years

If 2nd dose given,


a 4week interval
is required

Not required

2 doses if aged
14years

4weeks

Not required

* One of the doses should be given as dTpa (or dTpa-IPV if poliomyelitis vaccination is also
needed) and complete the course with dT. In the unlikely event that dT is not available,
dTpa or dTpa-IPV may be used for all 3 primary doses but this is not routinely recommended as
there are no data on the safety, immunogenicity or efficacy of dTpa for primary vaccination (see also
Chapter 3.14, Pertussis).
Varicella vaccine should be given to either non-immune people or people with no history
of previous varicella infection. At least 1 dose should be given to those aged <14years, and
all must receive 2 doses if aged 14years.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

38 The Australian Immunisation Handbook 9th Edition

1.4 Administration of vaccines


1.4.1Occupational health and safety issues
Standard occupational health and safety guidelines should always be followed
during a vaccination encounter to minimise the risk of needle-stick injury.1
Gloves are not routinely recommended for immunisation service providers. Work
practices should include the use of standard precautions to minimise exposure
to blood and body fluids. If exposure does occur, guidelines for post-exposure
prophylaxis should be followed (refer to Chapters 23 and 24 of the Australian
Government Department of Health and Ageing Infection control guidelines for the
prevention of transmission of infectious diseases in the health care setting).1
A new, sterile, disposable syringe and needle must be used for each injection.
Disposable needles and syringes must be discarded into a clearly labelled,
puncture-proof, spill-proof container that meets Australian standards in order to
prevent needle-stick injury or re-use.1 Always keep sharps containers out of the
reach of children. All immunisation service providers should be familiar with
the handling and disposal of sharps according to the Australian Government
Department of Health and Ageing Infection control guidelines for the prevention of
transmission of infectious diseases in the health care setting, Chapters 14 and 23.1

1.4.2 Equipment for vaccination


Preparing for vaccination
Depending on the vaccine(s) that are to be administered, and the age and size of
the person to be vaccinated, decide on the appropriate injection site and route,
and the injection equipment required (ie. syringe size, needle length and gauge).
The equipment chosen will vary depending on whether the vaccine is a
reconstituted vaccine, a vaccine from an ampoule or vial, or a vaccine in a
pre-filled syringe.

Equipment may include:


medical waste (sharps) container,
vaccine, plus diluent if reconstitution is required,
appropriate drawing-up needle (19 or 21 gauge needle if required, to draw
up through rubber bung and for reconstitution of vaccine),
appropriate injecting needle (see Table 1.4.2 Recommended needle size, length
and angle for administering vaccines),
clean cotton wool and hypoallergenic tape to apply to injection site after
vaccination, and
a rattle or noisy toy for distraction after the injection.

Administration of vaccines 39

1.4 Administration
of vaccines

2 or 3mL syringe (unless vaccine is in pre-filled syringe),

Preparing the vaccine


Wash hands carefully and prepare the appropriate injection equipment for
the vaccine to be administered.
Ensure that the minimum/maximum thermometer displays temperatures
within the +2C to +8C range before removing vaccine from the refrigerator.
Ensure that the correct vaccine is taken from the refrigerator and that it is within
the expiry date.
Check that there is no particulate matter or colour change in the vaccine.
Ensure that the diluent container is not damaged and potentially
contaminated.
PRECAUTIONS:
If a pre-filled syringe is provided, check carefully whether reconstitution
with vaccine (provided in a separate vial) is required.
The diluent of one brand of oral rotavirus vaccine (Rotarix) is provided
in a syringe-like oral plunger. Do not administer this vaccine by injection
(parenteral) after reconstitution.
Both rotavirus vaccines are administered orally.

Preparing vaccine provided in a pre-filled syringe, ampoule or liquid vial


If the vaccine is in a vial, remove the cap carefully to maintain sterility of the
rubber bung. Do not wipe the rubber bung. Use a 19 or 21 gauge needle to
draw up the recommended dose through the bung.
If the vaccine is in an ampoule, use a 23 gauge, 25mm needle to draw up the
recommended dose.
Needles should be changed after drawing up from a vial with a rubber bung,
but it is not necessary to change needles between drawing up a vaccine from
an ampoule and giving the injection.
Small air bubbles do not need to be extruded through the needle.
A needle or syringe that has already been used to inject an individual must
never come into contact with the vial because of the risk of cross-contamination.

40 The Australian Immunisation Handbook 9th Edition

Preparing vaccines requiring reconstitution


Reconstitute the vaccine as needed immediately before administration.
Never mix other vaccines together in the one syringe (unless that is the
manufacturers registered recommendation, eg. Infanrix hexa).
Never mix a local anaesthetic with a vaccine.
A sterile 21 gauge needle should be used for reconstitution and a separate 23
or 25 gauge needle, 25mm in length, should be used for administration of the
vaccine in most circumstances.
Use only the diluent supplied with the vaccine; do not use sterile water for
injection instead of a supplied diluent. Ensure that the diluent and vaccine
are completely mixed.
Reconstituted vaccines should be checked for signs of deterioration, such as a
change in colour or clarity.
Reconstituted vaccines may deteriorate rapidly and, in general, should be
administered as soon as practicable after they have been reconstituted.
Never freeze a vaccine after it has been reconstituted.

1.4.3 Route of administration


Almost all vaccines are given by either IM or SC injection, and a few vaccines
are given orally. Rotavirus vaccines are only available for oral administration
and must never be injected. Special training is required for intradermal
administration, which is important for several vaccines (see Chapter 3.17, Q
fever and Chapter 3.22, Tuberculosis). Table 1.4.1 below summarises the route of
administration for vaccines commonly used in Australia.

1.4 Administration
of vaccines

Administration of vaccines 41

Table 1.4.1: Route of administration for vaccines commonly used in Australia


Intramuscular
(IM) injection

Subcutaneous (SC)
injection

IM or SC injection

Oral

Diphtheria, tetanus
vaccine (dT)

Inactivated polio
vaccine (IPV)*

Influenza vaccine

Rotavirus vaccine
Cholera vaccine

Diphtheria,
tetanus, acellular
pertussis vaccine
(DTPa and dTpa)

Meningcoccal
polysaccharide
vaccine (4vMenPV)

Measles, mumps,
rubella vaccine
(MMR)

DTPa- and dTpacombination


vaccines
Hepatitis A vaccine
Hepatitis B vaccine
Hepatitis B
combination
vaccines

Varicella
vaccine (VV)
Q fever vaccine
Japanese
encephalitis vaccine
Measles, mumps,
rubella, varicella
vaccine (MMRV)
(when available)

Typhoid vaccine

Rubella vaccine
23-valent
pneumococcal
polysaccharide
vaccine (23vPPV)
Rabies vaccine
(HDCV)
Yellow fever vaccine

Haemophilus
influenzae type b
(Hib) vaccine
Human
papillomavirus
vaccine (HPV)
IPV-containing
combination
vaccines*
7-valent
pneumococcal
conjugate vaccine
(7vPCV)
Typhoid Vi
polysaccharide
vaccine
Meningococcal C
conjugate vaccine
(MenCCV)
Rabies vaccine
(PCECV)
* IPV-containing combination vaccines are administered by IM injection; IPV (IPOL) is administered by SC
injection.
The IM route is preferred because it causes fewer local adverse events.2
Q fever vaccine should be administered only by specially trained immunisation service providers.

42 The Australian Immunisation Handbook 9th Edition

1.4.4 Preparation for vaccine administration


Skin cleaning
Provided the skin is visibly clean, there is no need to wipe it with an antiseptic
(eg. alcohol wipe).3,4 If the immunisation service provider decides to clean the
skin, or if the skin is visibly not clean, alcohol and other disinfecting agents
must be allowed to dry before vaccine injection (otherwise there may be some
increased injection pain).

Distraction techniques
The routine use of distraction, relaxation and other measures have been
shown to reduce distress and pain following vaccination in young children.5-8
Reducing infant distress may enhance parents timely attendance for subsequent
vaccinations.
Distraction measures that may decrease discomfort following vaccination in young
children include:5-8
swaddling and holding the infant securely (but not excessively),
shaking a noisy toy (for infants and very young children),
playing music,
encouraging an older child to pretend to blow away the pain using a windmill
toy or bubbles, or
administering sweet-tasting fluid orally immediately before the injection
(with parental consent). In infants, 1520% sucrose drops have been used.
Topical anaesthetic agents, including vapocoolant sprays, are available but to
be effective must be applied at the correct time before vaccine administration.
Topical anaesthetics, such as EMLA, are not recommended for routine use,
but could be considered in a child with excessive fear or dislike of needles,
and require application 30 to 60 minutes before an injection.9 Vapocoolant
sprays are applied 15 seconds before vaccination. Topical lignocaine/prilocaine
is not recommended for children younger than 6months due to the risk of
methaemoglobinaemia.5

1.4 Administration
of vaccines

Administration of vaccines 43

1.4.5 Vaccine injection techniques


IM injection technique10,11
For IM injection, a 25mm needle should be used in most cases
(see Table 1.4.2 below).
Depending on the injection site, the limb should be positioned so as to relax
the muscle into which the vaccine is to be injected.
The 25mm needle should pierce the skin at an angle of 90 to the skin, and
can be safely inserted to the hub.12 Provided an injection angle of >70 is
used, the needle should reach the muscle layer.13
Studies have demonstrated that, for most vaccines, local adverse events
are minimised and immunogenicity enhanced by ensuring vaccine is
deposited into the muscle and not into the subcutaneous layer.5,14-17 However,
some vaccines, eg. inactivated poliomyelitis, varicella and meningococcal
polysaccharide vaccines, are only licensed for SC administration.
A recent clinical trial demonstrated that long (25mm) needles (with the skin
stretched flat and the needle inserted at 90) for infant vaccination were
associated with significantly fewer local adverse events while achieving
comparable immunogenicity. Little difference was found between needles
the same length but with different gauges in local adverse events or
immune response.12
If using a 25 gauge needle for an IM vaccination, ensure the vaccine is injected
slowly over a count of 5 seconds to avoid injection pain and muscle trauma.
It is not considered necessary to draw back on the syringe plunger before
injecting a vaccine.5 However, if this is done, and a flash of blood appears
in the needle hub, the needle should be withdrawn and a new site selected
for injection.18
After completing the injection, perform post-vaccination care
(see Chapter 1.5, Post-vaccination procedures).

SC injection technique
SC injections are usually administered at a 45 angle to the skin.
The standard needle for administering vaccines by SC injection is a 25 or 26
gauge needle, 16mm in length.

Intradermal injection technique


For intradermal injection of BCG vaccine or Q fever skin test vaccine, a 26 or
27 gauge, 10mm needle is recommended. The intradermal injection technique
requires special training, and should be performed only by a trained provider
(see Chapter 3.22, Tuberculosis and Chapter 3.17, Qfever).

44 The Australian Immunisation Handbook 9th Edition

Table 1.4.2: Recommended needle size, length and angle for administering
vaccines5,10,12,14,19
Age or size of child/adult

Needle type

Angle of needle insertion

Infant, child or adult


for IM vaccines

23 or 25 gauge,*
25mm in length

90 to skin plane

Preterm babies (<37weeks


gestation) up to age
2months; very small infants

23 or 25 gauge,*
16mm in length

90 to skin plane

Very large or obese patient

23 gauge, 38mm in length

90 to skin plane

Subcutaneous injection
in all individuals

25 or 26 gauge,
16mm in length

45 to skin plane

* If using a narrow 25 gauge needle for an IM vaccination, ensure vaccine is injected slowly
over a count of 5 seconds to avoid injection pain and muscle trauma.
The use of short needles for administering IM vaccines may lead to inadvertent
subcutaneous (SC) injection and increase the risk of significant local adverse events,
particularly with aluminium-adjuvanted vaccines (eg. hepatitis B vaccine, DTPa, DTPacombinations or tetanus vaccine).

1.4.6 Recommended injection sites


The choice of injection sites depends primarily upon the age of the individual
being vaccinated. The 2 anatomical sites recommended as routine injection sites
are the anterolateral thigh (Figure 1.4.6) and the deltoid muscle (Figure 1.4.9).
All practitioners should ensure that they are familiar with the landmarks used
to identify any anatomical sites used for vaccination. Photographs and diagrams
are provided in this section but are not a substitute for training. Further detail on
identifying the recommended injection sites is provided in Section 1.4.8.

Infants <12months of age


The vastus lateralis muscle in the anterolateral thigh is the recommended site
for IM vaccination in infants <12months of age (see Figures 1.4.5 and 1.4.6,
Section1.4.8).

The deltoid muscle is not recommended for IM vaccination of infants <12months


of age.

Children 12months of age


The deltoid muscle is the recommended site for IM vaccination in children
12months of age (see Figure 1.4.9, Section 1.4.8).

Administration of vaccines 45

1.4 Administration
of vaccines

The ventrogluteal area (see Figures 1.4.7 and 1.4.8, Section 1.4.8) is an alternative
site for IM vaccination of infants. It is important that vaccine providers who
choose to use this site are familiar with the landmarks used to identify it. The
reactogenicity and immunogenicity of vaccines given in this site are comparable
to those of vaccines given in the anterolateral thigh.20-22

The ventrogluteal area is an alternative site for IM vaccination of children


12months of age. However, vaccine providers should be familiar with the
landmarks used to identify this site.
The vastus lateralis in the anterolateral thigh may also be used in children
12months of age, but if this site is used, the less locally reactogenic vaccines, eg.
MMR, should be given in the thigh.

Adolescents and adults


The deltoid muscle is the recommended site for IM vaccination in adolescents
and adults (see Figure 1.4.9, Section 1.4.8).
The anterolateral thigh can also be used in older children and adults.
The ventrogluteal area is an alternative injection site. However, vaccine providers
should be familiar with the landmarks used to identify this site.
PRECAUTION:
Vaccine injections should not be given in the dorsogluteal site or
upper outer quadrant of the buttock because of the possibility of a
suboptimal immune response.23,24 Immunoglobulin can be administered
intramuscularly into the upper outer quadrant of the buttock, but care must
be taken to ensure that the other quadrants are not used.

46 The Australian Immunisation Handbook 9th Edition

1.4.7 Positioning for vaccination


It is important that infants and children do not move during injection of vaccines.
However, excessive restraint can increase their fear and result in increased
muscle tension. The following section describes a variety of positions which may
be used for vaccinating different age groups.

Positioning of infants <12months of age


Cuddle position for infants
Position the infant in a semi-recumbent cuddle position on the lap of the
parent/carer. The infants arm adjacent to the parent/carer should be restrained
underneath the parent/carers arm or against the parent/ carers chest. The
knee should be flexed to encourage relaxation of the vastus lateralis for IM
vaccinations. The infants other arm must also be held securely (see Figure 1.4.1).
This position can also be used for young children.
Figure 1.4.1: The cuddle position for vaccination of a child <12months of age

1.4 Administration
of vaccines

Photo courtesy Dr Joanne Molloy, VIC

Administration of vaccines 47

Positioning infant on an examination table


An alternative is to lay infants on their backs on an examination table, with the
infants feet towards the immunisation service provider, and the parent/carer
beside the provider to immobilise and distract the baby (see Figure 1.4.2).
Keep the infants hip and knee flexed by cupping the patella in the non-injecting
hand.
The thumb and index finger of the non-injecting hand may be used to stabilise
the hub of the needle once the needle has been inserted.
Figure 1.4.2: Positioning an infant on an examination table for vaccination

Photo courtesy Dr Joanne Molloy, VIC

Prone position across the lap for ventrogluteal vaccination


For ventrogluteal injection, position the child face-down across the parent/
carers lap. This allows the hips to be flexed and provides access to the
ventrogluteal area (see Figure 1.4.8).

48 The Australian Immunisation Handbook 9th Edition

Positioning of children 12months of age


Cuddle position for older child
Sit the child sideways on the lap of the parent/carer, with the arm to be injected
held close to the childs body while the other arm is tucked under the armpit and
behind the back of the parent/carer.
The childs exposed arm should be secured at the elbow by the parent/carer, and
the childs legs also secured by the parent/carer (see Figure 1.4.3).
Figure 1.4.3: Positioning an older child in the cuddle position

Photo courtesy Ann Kempe, MCRI, VIC

1.4 Administration
of vaccines

Administration of vaccines 49

Straddle position
An older child may be positioned facing the parent/carer with the legs straddled
over the parent/carers lap. The childs arms should be folded in front, with the
parent/carer hugging the childs body to the parent/carers chest. Alternatively,
the child may be positioned to hug the parent with the parents arms holding
the childs arms in a reciprocal hug (see Figure 1.4.4). This position allows access
to both deltoids and both anterolateral thighs.
Figure 1.4.4: Positioning a child in the straddle position

Photo courtesy Dr Joanne Molloy, VIC

50 The Australian Immunisation Handbook 9th Edition

Prone position across the lap for ventrogluteal vaccination


For ventrogluteal injection, position the child face-down across the parent/
carers lap (see Figure1.4.8).

Positioning of older children, adolescents and adults


Solo sitting position for deltoid injections
Most vaccines can be administered into the deltoid area. Adults should sit in
a straight-backed chair, feet resting flat on the floor with forearms and hands
in a relaxed position on the upper thighs. Keep the arms flexed at the elbow to
encourage the deltoid muscle to relax.
Encourage shoulders to drop by asking the person to raise the shoulders up while
taking a deep breath in and to drop them while breathing out fairly forcefully. Use
distraction to keep muscles relaxed during the procedure, eg. have an interesting
poster or similar for the person to concentrate on during the procedure and ask
him/her to give you a detailed description of what can be seen.
The ventrogluteal and vastus lateralis are alternative sites if needed (see above,
and below).

1.4.8 Identifying the injection site


The choice of injection site depends upon the age of the person, and is discussed
in Section 1.4.6.

The anterolateral thigh (vastus lateralis)


The infants nappy must be undone to ensure the injection site is completely
exposed and the anatomical markers easily identified.
Position the leg so that the hip and knee are flexed and the vastus lateralis is
relaxed (see Figure 1.4.6).
The upper anatomical marker is the midpoint between the anterior superior
iliac spine and the pubic tubercle, and the lower marker is the upper part of
the patella.
Draw an imaginary line between the 2 markers down the front of the thigh.
The correct site for IM vaccination is lateral to the midpoint of this line, in the
outer (anterolateral) aspect (see Figures 1.4.5 and 1.4.6).

Administration of vaccines 51

1.4 Administration
of vaccines

Do not inject into the anterior aspect of the thigh where neurovascular
structures can be damaged.

Figure 1.4.5: Diagram of the muscles of the thigh showing the anatomical
markers to identify the recommended (vastus lateralis) injection site (X)

Figure 1.4.6: Photograph of the thigh showing the recommended (vastus


lateralis) injection site (X)

Photo courtesy Lloyd Ellis, RCH, VIC

52 The Australian Immunisation Handbook 9th Edition

The ventrogluteal area


NB. This area should not be confused with the dorsogluteal area (buttock).
The ventrogluteal site provides an alternative site for administering vaccines
to a child of any age, especially when multiple injections at the same visit are
required. The ventrogluteal area is relatively free of major nerves and blood
vessels, and the area provides the greatest thickness of gluteal muscle.25,26 There is
a relatively consistent thinness of subcutaneous tissue over the injection site.26,27
The childs nappy must be undone to ensure the injection site is completely
exposed and the anatomical markers easily identified by sight and palpation.
Anatomical markers are the anterior superior iliac spine (ASIS), the greater
trochanter of the femur and the iliac crest (see Figure 1.4.7).
Place the child in a prone position (face-down) on parent/carers lap or on
the clinic table/bed with arms tucked against the childs chest. Allow the
childs legs to dangle towards the floor (see Figure 1.4.8).
The knee and hip should be turned inwards to encourage muscle relaxation at
the injection site.
The injection site should be that which is closest to the immunisation service
provider.
Place the palm over the greater trochanter (the uppermost bony prominence
of the thigh bone) with the thumb pointing towards the umbilicus. The
index finger points to the anterior superior iliac spine, and the middle finger
is spread so that it aims at the iliac crest, thus creating a V outlining the
ventrogluteal triangular area. The injection site is at the centre of this area
(see Figures 1.4.7 and 1.4.8).
Figure 1.4.7: Diagram showing the anatomical markers to identify the
ventrogluteal injection site (X) (ASIS = anterior superior iliac spine)

1.4 Administration
of vaccines

Administration of vaccines 53

Figure 1.4.8: Photograph with infant prone across carers lap, showing markers
to identify the ventrogluteal injection site (X)

Photo courtesy Dr Joanne Molloy, VIC

The deltoid area


It is essential to expose the arm completely from the top of the shoulder to the
elbow when locating the deltoid site (see Figure 1.4.9). Roll up the sleeve or
remove the shirt if needed.
The injection site is halfway between the shoulder tip (acromion) and the
muscle insertion at the middle of the humerus (deltoid tuberosity).
Draw an imaginary, inverted triangle below the shoulder tip, using the
identified anatomical markers.
The deltoid site for injection is the middle of the muscle (triangle)
(see Figure 1.4.9).

54 The Australian Immunisation Handbook 9th Edition

Figure 1.4.9: Diagram showing the anatomical markers to identify the deltoid
injection site

Subcutaneous injection sites


Subcutaneous injections should be administered either over the deltoid muscle
or over the anterolateral thigh. There are no data to demonstrate any difference
in technique between administration of a SC injection and a deep SC injection.
Figure 1.4.10 demonstrates the recommended technique for any SC injection.
Figure 1.4.10: A subcutaneous injection into the deltoid area of the upper arm
using a 25 gauge, 16mm needle, inserted at a 45 angle

1.4 Administration
of vaccines

Photo courtesy Ann Kempe, MCRI, VIC

Administration of vaccines 55

1.4.9 Administering multiple vaccine


injections at the same visit
The location of each separate injection given should be recorded, so that if a local
adverse event occurs, the implicated vaccine(s) can be identified.

Infants <12months of age


The suitable sites for this age group are the anterolateral thighs and the
ventrogluteal areas. Two vaccines can be given into the same anterolateral thigh,
separated by at least 2.5 cm. However, only 1 vaccine should be given into each
ventrogluteal area.
When 3 or 4 IM vaccines are to be given at the same visit, the options are:
2 injections can be administered in the same anterolateral thigh, separated by
at least 2.5 cm (see Figure 1.4.11); further IM vaccines can be given in this way
in the other thigh, or
1 injection can be given into each anterolateral thigh and 1 injection can be
administered into each ventrogluteal area.
Figure 1.4.11: Recommended technique for giving multiple vaccine injections
to an infant <12months of age into the anterolateral thigh

56 The Australian Immunisation Handbook 9th Edition

Children 12months of age, adolescents and adults


A single injection can be given into each deltoid muscle.
When 3 or 4 IM vaccines are to be given to a child at the same visit, the options
will depend on the muscle mass of the childs deltoid.
If the deltoid mass is adequate:
a further injection can be given into each deltoid muscle (separated by 2.5 cm
from the initial vaccine).
If the deltoid muscle mass is small:
further injections can be given into either the anterolateral thighs (2.5 cm
apart if 2 vaccines are given in the same thigh), or
give 1 injection into each ventrogluteal area.
For younger children, the cuddle or straddle position (Figures 1.4.3 and 1.4.4) are
suitable for accessing multiple limbs during the one vaccination encounter.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

1.4 Administration
of vaccines

Administration of vaccines 57

1.5 Post-vaccination procedures


1.5.1 Immediate after-care
Dispose of clinical waste, including sharps and vaccine vials, immediately
after administration of the vaccine and at its point of use. Refer to the State/
Territory health authority for management guidelines for the safe disposal of
clinical waste or refer to the Australian Government Department of Health
and Ageing Infection control guidelines for the prevention of transmission of
infectious diseases in the health care setting.1
Cover the site quickly with a dry cotton ball and tape as needed.
Gently apply pressure for 1 or 2 minutes. Do not rub the site as this will
encourage the vaccine to leak back up the needle track, which can cause pain
and may lead to local irritation.
Remove the cotton wool after a few minutes and leave the injection site
exposed to the air.
Paracetamol is not routinely used before or at the time of vaccination, but
may be recommended as required for fever or pain.
To distract the individual and reduce distress, immediately change the position
of the child/person after completing the vaccination, eg. ask the parent/carer
to put the infant over the shoulder and move around with the infant.2
The vaccinated person and/or parent/carer should be advised to remain in
a nearby area for a minimum of 15 minutes after the vaccination. The area
should be close enough to the immunisation service provider, so that the
individual can be observed and medical treatment rapidly provided if needed.
Take the opportunity to check the vaccination status of other family members
(as appropriate) and provide (or refer) for catch-up vaccination.
Record the relevant details of the vaccines given in a record to be retained
by the person or parent/carer, in the surgery/clinic record and, for children
aged <7years, forward records to the ACIR (see Section 1.5.3, Documentation
of vaccination).
Before departure, inform the individual or parent/carer, preferably in
writing, of the date of the next scheduled vaccinations.

1.5.2 Adverse events following immunisation


What are AEFI?
An adverse event following immunisation (AEFI) is an unwanted or unexpected
event occurring after the administration of vaccine(s). Such an event may be
caused by the vaccine(s) or may occur by chance after vaccination (ie. it would
have occurred regardless of vaccination). Most vaccines cause minor adverse
events such as low-grade fever, pain or redness at the injection site and these

58 The Australian Immunisation Handbook 9th Edition

The frequency of adverse events can be classified as follows: very common (>10%),
common (110%), uncommon (0.11%), rare (0.010.1%) and very rare (<0.01%).4

Common adverse events


The following common adverse events should be anticipated following
vaccination.5 They can be distressing for parents/carers, but they do not
contraindicate further vaccination. In general, unless these adverse events are
significant, they do not need to be reported by immunisation service providers
to the Adverse Drug Reactions Advisory Committee (ADRAC) (see Table 1.5.3,
Contact details for notification of AEFI).
Parents/carers should be given advice (preferably written) as part of the consent
procedure on what common adverse events are likely and what they should do
about them (the table inside the back cover of this Handbook, Commonly observed
adverse events following immunisation with vaccines used in the National Immunisation
Program (NIP) schedule and what to do about them, can be used for this purpose).
DTPa, dTpa, hepatitis B, Hib, IPV and their various combinations may cause
transient minor adverse events including swelling, redness or soreness at the
injection site, and low-grade fever, crying and irritability (in infants).
There is an increased risk of more extensive local adverse events after booster
doses of DTPa and DTPa-combination vaccines.6 A local adverse event that
involves extensive limb swelling should be reported. For the definition of
extensive limb swelling, see Appendix 6, Definitions of adverse events following
immunisation.
MMR vaccine may be followed 5 to 12days later by a fever lasting 2 or
3days, malaise and/or rash. This is not infectious. Fever >39.4C is very
common, occurring in 5 to 15% of vaccinees, 5 to 12days after vaccination.
Human papillomavirus vaccine may cause mild injection site adverse events
(pain, swelling and erythema) and, occasionally, headache, fever and nausea.
Influenza vaccine may cause soreness at the injection site. Fever, malaise, and
myalgia occur less commonly.
The 7vPCV causes low-grade fever and/or mild pain at the injection site in
about 10% of infant recipients. The 23-valent pneumococcal polysaccharide
vaccine (23vPPV) causes mild local adverse events in up to half the adult
recipients.
MenCCV is generally well tolerated. Very common (>10%) adverse events
are pain, redness and swelling at the injection site, fever, irritability, anorexia
and headache.

Post-vaccination procedures 59

1.5 Post-vaccination
procedures

should be anticipated3 (see the table Comparison of the effects of diseases and the side
effects of vaccines inside the front cover of this Handbook).

Varicella vaccine may cause mild local soreness and swelling. A mild
maculopapular or papulovesicular rash occurs in up to 5% of vaccinated
children (see also Chapter 3.24, Varicella).
Injection site nodules are not uncommon. They are fibrous remnants of the
bodys interaction with the vaccine components (usually an adjuvant) in the
muscle, and they may remain for manyweeks after the vaccination. Injection
site nodules do not require any specific treatment.
Oral rotavirus vaccine may cause mild fever and/or diarrhoea (see
Chapter3.18, Rotavirus).

Managing common adverse events


Advice to parents on common adverse events
Vaccine injections may result in soreness, redness, itching, swelling or burning
at the injection site for 1 to 2days. Paracetamol might be required to ease the
discomfort.

Managing fever after vaccination


Routine use of paracetamol at the time of vaccination is no longer recommended.
If an infant or child has a fever of >38.5C following vaccination, paracetamol can
be given. The dose of paracetamol is 15mg/kg/dose of paracetamol liquid, up to
a maximum daily dose of 90mg/kg per day in 4 to 6 divided doses for up to 48
hours.

Preventing AEFI
The key to preventing uncommon or rare adverse events is to screen each person
to be vaccinated using pre-vaccination screening (Tables 1.3.1 and 1.3.2) to
ensure that the person does not have a condition which either increases the risk
of an adverse event or is a contraindication to vaccination. The correct injection
technique is also important. Immunisation service providers should also check
the relevant chapters of this Handbook or the product information supplied
with the vaccine for more details on precautions and contraindications for each
vaccine they are to administer.

Uncommon and rare AEFI


Some vaccines have been shown to cause uncommon or rare adverse events,
although the rate is always hundreds to thousands times less frequent than the
disease complications. Examples are given below.

Rare, late events shown to be causally related to some vaccines


The use of oral poliomyelitis vaccine (OPV) in Australia was discontinued in
2005. OPV can rarely cause vaccine-associated paralytic poliomyelitis (VAPP).
The incidence is 1 in 2.4 million doses of OPV, which means that Australia would
have expected 1 case of VAPP every 3years when OPV was in use. However,

60 The Australian Immunisation Handbook 9th Edition

Vaccines containing diphtheria and tetanus have been described as causing


brachial neuritis, with an incidence of approximately 1 in 100000 (adults).

Events where evidence demonstrates no


causal link with immunisation
There is epidemiological evidence which indicates that there is no causal
association between immunisation and the following events:
sudden infant death syndrome (SIDS) and any vaccine,8-10
autism and MMR vaccine,11-14
multiple sclerosis and hepatitis B vaccine,15-18
inflammatory bowel disease and MMR vaccine,19
diabetes and Hib vaccine,20-22
asthma and any vaccine.23

Management of an immediate AEFI


Observation after vaccination
Recipients of vaccines should remain under observation for a short interval
to ensure that they do not experience an immediate adverse event. It is
recommended that recipients remain in the vicinity of the place of vaccination
for at least 15 minutes. Severe anaphylactic reactions usually have a rapid onset;
most life-threatening adverse events begin within 10 minutes of vaccination.
The most serious immediate AEFI is anaphylaxis. However, in adults and older
children, the most common immediate adverse event is a vasovagal episode
(fainting), either immediately or soon after vaccination. Because fainting after
vaccination can lead to serious consequences, anyone who complains of giddiness
or light-headedness before or after vaccination should be advised to lie down
until free of symptoms. Most faints following vaccination occur within 5 minutes,
and 98% occur within 30 minutes. Adults should, therefore, be warned of the risk
of driving or operating machinery for at least 30 minutes after vaccination.24
Children who have had a serious adverse event (other than a contraindication,
such as anaphylaxis) to a previous vaccine may subsequently be vaccinated under
close medical supervision. Check with State/Territory health authorities for more
information (see Section 2.3.1, Vaccination of children who have had a serious adverse
event following immunisation and Appendix 1, Contact details for Australian, State and
Territory Government health authorities and communicable disease control).

Anaphylaxis and vasovagal episodes


Anaphylaxis following routine vaccination is very rare, but can be fatal.
All immunisation service providers must be able to distinguish between
anaphylaxis, convulsions and fainting.

Post-vaccination procedures 61

1.5 Post-vaccination
procedures

the reported incidence of VAPP was only 1 case every 8 to 9years in Australia.7
VAPP does not occur from vaccination with IPV or IPV-containing vaccines.

Fainting (vasovagal episode) is relatively common after vaccination of adults and


adolescents, but infants and children rarely faint. Sudden loss of consciousness in
young children should be presumed to be an anaphylactic reaction, particularly
if a strong central pulse is absent. A strong central pulse (eg. carotid) persists
during a faint or convulsion.
The features listed in Table 1.5.1 may be useful in differentiating these 2conditions.
If the diagnosis is unclear and anaphylaxis is considered, management for this
should be instituted with the prompt administration of adrenaline.
Table 1.5.1: Clinical features which may assist differentiation between a
vasovagal episode and anaphylaxis
Vasovagal episode

Anaphylaxis

Immediate, usually within


minutes of or during
vaccine administration.

Usually within 15 minutes,


but can occur within hours,
of vaccine administration.

Skin

Generalised pallor,
cool, clammy skin.

Skin itchiness, generalised


skin erythema (redness),
urticaria (weals) or
angioedema (localised
oedema of the deeper
layers of the skin or
subcutaneous tissues).

Respiratory

Normal respiration; may be


shallow, but not laboured.

Cough, wheeze, stridor,


or signs of respiratory
distress (tachypnoea,
cyanosis, rib recession).

Cardiovascular

Bradycardia, weak/
absent peripheral pulse,
strong carotid pulse.

Tachycardia, weak/absent
peripheral and carotid pulse.

ONSET

Symptoms/
Signs

Hypotension usually
transient and corrects
in supine position.
Neurological

Feels faint, light-headed.


Loss of consciousness
improves once supine or
head down position.

Hypotension sustained
and no improvement
without specific treatment.
Sense of severe anxiety
and distress.
Loss of consciousness no
improvement once supine
or head down position.

Signs of anaphylaxis
Anaphylaxis is a severe adverse event of rapid onset, characterised by sudden
respiratory compromise and/or circulatory collapse. Early signs include
involvement of the skin, eg. generalised erythema, urticaria and/or angioedema
(swelling), and/or gastrointestinal tract, eg. diarrhoea, vomiting. In severe
cases, there is circulatory collapse with alteration in the level of consciousness,

62 The Australian Immunisation Handbook 9th Edition

Immunisation service providers should be able to recognise all the following


symptoms and signs of anaphylaxis:
cutaneous, such as the rapid development of widespread urticarial lesions
(circumscribed, intensely itchy weals with erythematous, raised edges and
pale, blanched centres) and/or erythema and/or angioedema (soft tissue
swelling usually affecting the face and/or limbs),
upper airway obstruction, such as hoarseness and stridor, resulting from
angioedema of the hypopharynx, epiglottis and larynx,
lower airway obstruction, such as subjective feelings of retrosternal tightness,
and dyspnoea with audible expiratory wheeze from bronchospasm,
limpness and pallor, which are signs of hypotension in infants and young
children,
profound hypotension in association with other signs of cardiovascular
disturbance, such as sinus tachycardia or severe bradycardia, absent central
pulses and reduced peripheral circulation, and/or
abdominal cramps, diarrhoea and/or vomiting.

Management of anaphylaxis
Rapid IM administration of adrenaline is the cornerstone of treatment of
anaphylaxis.
Anaphylaxis occurs without warning, usually within 15 minutes of giving a
vaccine. A protocol for the management of anaphylaxis, adrenaline, and 1mL
syringes must always be immediately at hand whenever vaccines are given.
If the patient is unconscious, lie him/her on the left side and position to keep
the airway clear. If the patient is conscious, lie supine in head down and feet
up position (unless this results in breathing difficulties).
Give adrenaline by IM injection (see below for dosage) for any signs of
anaphylaxis with respiratory and/or cardiovascular symptoms or signs.
Adrenaline is not required for generalised non-anaphylactic reactions (such
as skin rash or angioedema). If in doubt, IM adrenaline should be given.
If there is no improvement in the patients condition by 5 minutes, repeat
doses of adrenaline every 5 minutes until improvement occurs.
If oxygen is available, administer by facemask at a high flow rate.
Call for assistance. Never leave the patient alone.
Begin expired air resuscitation for apnoea, check for a central pulse. If pulse
is not palpable, commence external cardiac massage (ECM).
All cases should be admitted to hospital for further observation and
treatment.

Post-vaccination procedures 63

1.5 Post-vaccination
procedures

hypotension and weak or absent pulses, and/or marked respiratory compromise


from upper airway oedema or bronchospasm.

Document the time and dose of adrenaline given.


Experienced practitioners may choose to use an oral airway if the appropriate
size is available, but its use is not routinely recommended unless the patient is
unconscious.
Antihistamines and/or hydrocortisone are not recommended for the emergency
management of anaphylaxis.

Adrenaline dose
Adrenaline 1:1000 (one in one thousand)
Adrenaline 1:1000 contains 1mg of adrenaline permL of solution in a 1mL
glass vial. Adrenaline 1 in 10000 is no longer recommended for the treatment
of anaphylaxis. The use of 1:1000 adrenaline is recommended because it
is universally available. Use a 1mL syringe to improve the accuracy of
measurement when drawing up small doses.
The recommended dose of 1:1000 adrenaline is 0.01mL/kg body weight
(equivalent to 0.01mg/kg or 10g/kg) up to a maximum of 0.5mL, given by
deep IM injection (not the deltoid). Adrenaline 1:1000 must not be administered
intravenously. Table 1.5.2 lists the dose of 1:1000 adrenaline to be used if the exact
weight of the individual is not known.
Table 1.5.2: Doses of intramuscular 1:1000 (one in one thousand) adrenaline for
anaphylaxis
Less than 1 year

0.050.1mL

12years (approx. 10kg)

0.1mL

23years (approx. 15kg)

0.15mL

46years (approx. 20kg)

0.2mL

710years (approx. 30kg)

0.3mL

1112years (approx. 40kg)

0.4mL

13years and over (over 40kg)

0.5mL

The dose of 1:1000 (one in one thousand) adrenaline may be repeated every 5
minutes as necessary until there is clinical improvement.

Reporting AEFI
Surveillance for adverse events following immunisation is an integral part of a
national vaccination program. Through surveillance, it is hoped to detect changes
in the rates of known adverse events and any adverse events that either were
previously undocumented, or result from program errors, such as incorrect
vaccine schedule, delivery or storage.

64 The Australian Immunisation Handbook 9th Edition

Any of the adverse events listed in Appendix 6, Definitions of adverse events following
immunisation should be reported. No time limit has been set to report AEFI.
Notification of an adverse event does not necessarily imply a causal association
with vaccination, as some events may occur coincidentally following vaccination.
Immunisation service providers are also advised to report any adverse events
of concern that do not fit into any of the categories listed in Appendix 6. They
should be reported as other reactions with a full description of the adverse
event. This will enable new and unexpected AEFI to be identified.

How should AEFI be reported?


AEFI are notifiable directly to the relevant health authority in Australian Capital
Territory, New South Wales, Northern Territory, Queensland, South Australia,
Victoria and Western Australia. In Tasmania, AEFI should be reported using the
Adverse Drug Reactions Advisory Committee (ADRAC) blue card.
AEFI are notifiable conditions in Australian Capital Territory, New South Wales,
Northern Territory, Queensland, South Australia, Victoria and Western Australia
and must be reported directly to the relevant health authority (see Table1.5.3
below). These State and Territory health authorities then forward AEFI
notifications to ADRAC.
The Adverse Drug Reactions Advisory Committee (ADRAC) receives reports
of unexpected and serious adverse events for all medicines, including
vaccines. Any person (medical or non-medical) can report an AEFI to ADRAC
by telephoning the numbers listed in Table 1.5.3 below, or by filling in a blue
card or completing a web-based report (https://www.tgasime.health.gov.au/
SIME/ADRS/ADRSLodg.nsf/wNotification?OpenForm).
Additional blue cards are available from:
The Secretary
Adverse Drug Reactions Advisory Committee
PO Box 100
Woden ACT 2606
Telephone: 1800 044 114 or on-line at www.tga.gov.au/adr/bluecard.htm
ADRAC will forward copies of individual reports of AEFI with vaccines on the
National Immunisation Program schedule to those States/Territories that have
follow-up surveillance. In addition, reports from ADRAC and State/Territory
Health Departments are aggregated and published in Communicable Diseases
Intelligence.25

Post-vaccination procedures 65

1.5 Post-vaccination
procedures

Any serious or unexpected adverse event following immunisation should


be reported. Providers should use clinical judgement and common sense
in deciding which adverse events to report, and parents/carers should
be encouraged to notify the immunisation service provider or health
authorities of an AEFI.

Table 1.5.3: Contact details for notification of AEFI


State/Territory

Report adverse events


directly to:

Telephone number

*Australian
Capital Territory

ACT Health Department

02 6205 2300

*New South Wales

NSW Public Health Units

Contact your local Public Health Unit,


found under Health in the White Pages

*Northern Territory NT Department of Health


and Community Services

08 8922 8044

*Queensland

Queensland Health

07 3234 1500

*South Australia

Department of Health

08 8226 7177

In SA, parents can also report


adverse events by calling

1300 364 100 (24 hours)

Tasmania

ADRAC

Use blue card

*Victoria

Department of Human
Services, SAEFVIC

1300 822 924

*Western Australia

State Health Department

08 9321 1312

* AEFI are notifiable in these States/Territories and health professionals should report
directly to their respective Health Department as listed above.

1.5.3 Documentation of vaccination


A personal health record should be established for each vaccinee and newborn
infant, and kept by that person or the parent/carer. The parent/carer should be
urged to present the record every time a child is seen by a health professional.
The following details should be recorded in the personal health record, and in the
clinical file:
the vaccinees full name and date of birth,
the details of the vaccine given, including the dose, brand name, batch
number, and site of administration,
the name of the person providing the vaccination,
the date of vaccination, and
the date the next vaccination is due.
If the vaccinee is a child <7years of age, the Australian Childhood Immunisation
Register (ACIR) must also be notified of the vaccination details (see The
Australian Childhood Immunisation Register below).

66 The Australian Immunisation Handbook 9th Edition

1.5.4 The Australian Childhood Immunisation Register

Children enrolled in Medicare are automatically included on the ACIR. Children


not enrolled in Medicare will be included when an immunisation service
provider sends details of a vaccination to the ACIR. No vaccination information
is recorded on the ACIR after a child turns 7years of age, but any information
already held is retained. The information will relate only to vaccines received
between the ages of birth and the 7th birthday. The ACIR Enquiry Line can be
contacted on 1800653809 (free call) and any record held for an individual who is
7years of age can also be made available to an immunisation service provider
or parent/carer.
The ACIR provides an important means of accountability and evaluation of
the childhood vaccination program. It is the primary means of determining
vaccination coverage at national, State/Territory and local levels. It also provides
a central vaccination history for each child that is accessible to any Australian
immunisation service provider wishing to assess vaccination status. Since 1998,
data held on the ACIR have been used to determine a familys entitlement to the
Child Care Benefit and Maternity Immunisation Allowance family assistance
payments. It is, therefore, important that vaccination data are submitted to the
ACIR promptly.

Reporting to ACIR
Immunisation service providers should send to the ACIR details of all NIP and
private vaccinations given to children <7years of age. Vaccination details may
be submitted by sending data electronically via Medicare Australias on-line
claiming facility, Electronic Data Interchange (EDI) on the Internet, or by using
a paper form. Providers in Queensland and the Northern Territory currently
sending data to the ACIR via their State/Territory Health Department should
continue to do so. Providers in all other States/Territories should send data
directly to the ACIR.
A childs vaccination record can also be updated with vaccination details where
the vaccination was performed by another immunisation service provider,
including those given while the child was overseas, by completing and sending
an Immunisation History form to Medicare Australia. Forms are available on the
ACIR website at www.medicareaustralia.gov.au/providers/forms/acir.htm.

Post-vaccination procedures 67

1.5 Post-vaccination
procedures

The Australian Childhood Immunisation Register (ACIR) is a national database


for recording details of vaccinations given to children <7years of age who live
in Australia. It commenced on 1 January 1996 and is administered by Medicare
Australia under the legislative mandate of the Commonwealth Health Insurance
Act 1973 Part IVA. Section 46B of the Health Insurance Act specifies how the ACIR
is to be implemented and managed. Section 46E sets out the provisions for giving
both de-identified and identified information to recognised immunisation service
providers and other specified agencies.

When relevant, immunisation service providers should complete the Conscientious


Objection and Medical Contraindication forms and forward to the ACIR.
For further information about the ACIR and reporting vaccination information,
see The ACIR Internet site below. In addition, assistance on any reporting issues
can be obtained from the ACIR Enquiry Line, 1800653809 (free call).

Immunisation History Statement


Immunisation History Statements, which contain details of all vaccines
administered to the child and recorded on the ACIR, and those that may be
missing, are automatically generated when a child turns 12months, 2 and 5years
of age and on completion of the childhood vaccination schedule. Statements will be
mailed to the address most recently recorded on the ACIR for that child.
Parent/carers can also get a Statement at any other time:
on-line at www.medicareaustralia.gov.au,
from their local Medicare office,
by calling 1800 653 809 (free call).
Immunisation History Statements can be used when proof of vaccination is
needed. For example, Statements can be used to meet vaccination requirements for:
primary school enrolment a sentence will be displayed at the bottom of the
statement that says the child has received all the vaccinations required by
5years of age, and/or
eligibility for the Child Care Benefit and Maternity Immunisation Allowance;
an up-to-date status for the Family Assistance Office will be displayed.

Recording details of a deceased child


The ACIR should be notified of a deceased child to prevent an Immunisation
History Statement being sent to bereaved parents/carers. Advice of a childs
death can be provided by calling 1800 653 809 (free call), or by sending details
on practice stationery. Details should include the childs name, address, date of
birth, Medicare number and date of death.

Children who have moved to live overseas


A child who has moved overseas can be removed from the ACIR by sending
details to the ACIR by fax, phone or secure site email. This prevents the childs
name continuing to appear on ACIR reports of overdue children.

Ascertaining individual vaccination status


Parents/carers can telephone the ACIR on 1800 653 809 (free call) for information
about their childs vaccination status, regardless of where the childs vaccination
was given. Immunisation service providers can also request a childs vaccination
status by telephone.

68 The Australian Immunisation Handbook 9th Edition

Vaccination coverage and other reports

Practices that are registered for the General Practice Immunisation Incentive
(GPII) scheme can receive quarterly reports on vaccination coverage for children
within that practice. Other reports, including those that identify a childs
vaccinations and due/overdue details, are available through the secure area of
the ACIR Internet site to approved immunisation service providers.

The ACIR Internet site


The ACIR Internet site has 2 main parts, a general information area and a secure
area. The Internet address for the ACIR is www.medicareaustralia.gov.au. Any
person with Internet access may view the ACIR site for general vaccination
information and statistics.
Approved immunisation service providers are able to access the secure area of
the ACIR Internet site and obtain a range of statistical and identified reports.
These reports are available, depending on the access level granted to the provider,
and enable approved providers to view a childs vaccination details, record
vaccination information and access a range of other reports. To register for access
to the secure area of the ACIR Internet site, providers should complete the online
request form at http://www.medicareaustralia.gov.au/providers/programs_
services/acir/index.htm. Further information or assistance may be obtained by
calling the ACIR Internet Helpline on 1300 650 039 (free call).

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

Post-vaccination procedures 69

1.5 Post-vaccination
procedures

ACIR reports assess progress towards national targets, and help to identify areas
with low vaccination levels and assist in planning vaccination programs.

PART 2: Vaccination for


special risk groups
2.1 Vaccination for Aboriginal and
Torres Strait Islander people
Aboriginal and Torres Strait Islander people historically had a very high
burden of infectious diseases, including those for which vaccines were
subsequently developed. These high rates of disease in the early period of
European colonisation were mainly due to a lack of previous exposure and
acquired immunity,1 and in more recentyears have been associated with lower
standards of living and poorer access to water, housing and health care.2 For
some vaccine-preventable diseases (VPDs), such as diphtheria, polio, tetanus,
hepatitis B, measles, mumps and rubella, vaccination has been very successful in
eliminating or substantially reducing disease in all Australians, and has made a
substantial contribution to improvements in Aboriginal and Torres Strait Islander
child mortality in recent decades.3 For some other VPDs, in particular invasive
pneumococcal disease and influenza in adults, greater burdens of illness still
occur in Indigenous compared to non-Indigenous people and remain a major
cause of illness and death.3 Detailed information on the current status of VPDs
and vaccination status in Aboriginal and Torres Strait Islander people is available
elsewhere.3
This chapter discusses the vaccines for which there are different
recommendations for Aboriginal and Torres Strait Islander people in at least
some parts of Australia. These are, for children, BCG, hepatitis A, Haemophilus
influenzae type b, and pneumococcal vaccines, and, for adults, influenza and
pneumococcal polysaccharide vaccines.

CHILDREN
BCG vaccine and tuberculosis
In the past, Aboriginal and Torres Strait Islander people have suffered from
much higher rates of tuberculosis, more than 20 times the rate of non-Indigenous
Australian-born people in some areas.4 While there have been substantial
improvements in disease rates in recent decades, tuberculosis remains more
common in Indigenous than non-Indigenous Australian-born people in many
parts of Australia, particularly northern and central Australia.5,6 Although there
is uncertainty about the efficacy of BCG in preventing pulmonary tuberculosis,
it provides substantial protection against disseminated forms of the disease
in young children.7 BCG is therefore recommended for Aboriginal and Torres
Strait Islander neonates in regions of high incidence (Northern Territory, far
northern Queensland, some regions of both Western Australia and South

70 The Australian Immunisation Handbook 9th Edition

Haemophilus influenzae type b


Before the introduction of an effective Haemophilus influenzae type b (Hib)
vaccine, not only was the incidence of invasive Hib disease very high in
Aboriginal and Torres Strait Islander children, particularly in more remote
areas, it also occurred at a younger age than in non-Indigenous children. Thus, a
vaccine to prevent Hib disease in Aboriginal and Torres Strait Islander children
needed to be immunogenic as early as possible in infancy. The vaccine known
by the abbreviation PRP-OMP (PedvaxHIB or COMVAX) is more immunogenic
at 2months of age than the other conjugate Hib vaccines, and so was the
preferred Hib vaccine for Aboriginal and Torres Strait Islander children from
the inception of the Hib vaccination programs in 1993. Since then, there has
been a dramatic decline of Hib disease in Aboriginal and Torres Strait Islander
children.10,11 The experience in other high incidence populations indicates that
it is important to continue to use PRP-OMP vaccine in Aboriginal and Torres
Strait Islander populations demonstrated to be at highest risk, as in central and
northern Australia. However, the available data indicate that Indigenous children
in areas of low incidence, and non-Indigenous children, have a pattern of Hib
disease which is adequately covered by a vaccine not prompting significant
immune response until after the second dose.12 New combination vaccines
which include a Hib (PRP-T) component have the advantage of reducing the
number of injections required. Therefore, in the Northern Territory, Queensland,
South Australia and Western Australia, all Aboriginal and Torres Strait Islander
children should receive a Hib vaccine with a PRP-OMP component, while
Indigenous children in other jurisdictions, and non-Indigenous children, may
receive either PRP-T or PRP-OMP Hib vaccines (see Chapter 3.4, Haemophilus
influenzae type b). State/Territory health authorities should be contacted about the
vaccination schedule for each jurisdiction.

Vaccination for Aboriginal and Torres Strait Islander people 71

2.1Vaccination for
Aboriginal and Torres
Strait Islander people

Australia), where infants are at higher risk of acquiring this serious, and often
fatal, condition. State and Territory guidelines should be consulted where BCG is
being considered for neonates <2.5kg in weight. Nevertheless, as the incidence
of pulmonary tuberculosis in adults and the risk of disseminated tuberculosis in
infants decreases, the risk of severe complications of BCG, documented in native
peoples elsewhere, may become a significant consideration.8 State/Territory
health authorities should be consulted to determine the recommendations for
particular areas. It is usually administered to eligible infants by hospital staff
(ie. midwives or nurses who have been specially trained) soon after delivery.
Injection technique is particularly important for BCG vaccination which must be
administered intradermally. Adverse events, such as regional lymphadenitis, are
less common when administration is performed by trained staff.9 See Chapter
3.22, Tuberculosis for more information.

Hepatitis A
Hepatitis A infection has been shown to be very common in Aboriginal and
Torres Strait Islander children across northern Australia.13-15 Although the
symptoms of infection in early childhood are usually mild or absent, cases
complicated by liver failure and death have been reported among Indigenous
children in far north Queensland15 and the Kimberley13 and recorded
hospitalisation rates have been found to be at least 50 times higher in Indigenous
compared to non-Indigenous children.3 A vaccination program for Indigenous
children was introduced in north Queensland in 1999, which resulted in
substantial decreases in disease rates not only in Indigenous but also in nonIndigenous children, suggesting a substantial herd immunity effect.16 Vaccination
is now recommended for Aboriginal and Torres Strait Islander children in those
jurisdictions with high incidence: the Northern Territory, Queensland, South
Australia and Western Australia (see Chapter 3.5, Hepatitis A). Two doses should
be given, commencing in the second year of life. As the exact recommended
ages of administration vary between States and Territories, jurisdictional health
authorities should be contacted about their vaccination schedules.

Pneumococcal vaccines
Some of the highest rates of invasive pneumococcal disease (IPD) ever reported
in the world were in young central Australian Aboriginal children before the
availability of the conjugate vaccine,17 and very high rates were also reported
in Indigenous children in other parts of northern Australia.18,19 High rates of
pneumococcal pneumonia have also been documented in central Australian
children,20 and Streptococcus pneumoniae has been implicated in the high rates of
otitis media.21 In response to this, the 7-valent pneumococcal conjugate vaccine
(7vPCV) was made available for Aboriginal and Torres Strait Islander children
from 2001. As well as higher rates of IPD, a wider range of serotypes is responsible
for disease in Aboriginal and Torres Strait Islander children, resulting in a lower
percentage of cases (below 60%) caused by serotypes included in the 7vPCV.18,19
Therefore, a booster dose of 23-valent pneumococcal polysaccharide vaccine at
1824months of age, following the primary course of 7vPCV, is recommended in
areas of high incidence, ie. the Northern Territory, Queensland, South Australia
and Western Australia. See Chapter 3.15, Pneumococcal disease for more information.
There has been a rapid decline in invasive pneumococcal disease in Indigenous
children since the introduction of the pneumococcal vaccines in 2001.22

ADULTS
Influenza
Influenza and/or pneumonia is the primary cause of around 2.5% of deaths in
Aboriginal and Torres Strait Islander people, the vast majority being adults.2
The disease burden is greatest in the elderly, with hospitalisation and death
more than twice as frequent in Indigenous adults aged 50years, compared

72 The Australian Immunisation Handbook 9th Edition

to non-Indigenous adults.3 Younger Indigenous adults suffer an even greater


relative burden than non-Indigenous younger adults, at least 7 times higher
for hospitalisations, and 28 times higher for death,3 probably related to a high
prevalence of risk factors such as diabetes, renal disease and excessive alcohol use.2
The most common complication of influenza is secondary bacterial pneumonia,
and influenza vaccine has been shown to be effective in preventing pneumonia
and death in the elderly.23 Therefore, yearly influenza vaccination is
recommended for all Aboriginal and Torres Strait Islander adults aged 15years.

Pneumococcal polysaccharide vaccine

23vPPV is recommended for all Aboriginal and Torres Strait Islander people
aged 50years, and for those aged 1549years who have high-risk underlying
conditions, and has been funded nationally for people in these categories since
1999. Eligibility for Indigenous adults may be broader than this in some regions;
jurisdictional health authorities should be contacted for further information. A
single revaccination is recommended after 5years, and a second revaccination is
recommended in some circumstances. See Chapter 3.15, Pneumococcal disease for
more details.

Other vaccines
The first ever outbreak of Japanese encephalitis (JE) in Australia occurred in the
remote outer islands of the Torres Strait in 1995. JE vaccine was first offered to
the residents of these islands in late 1995 and, since then, the vaccine has been
integrated into the childhood vaccination schedule commencing at 12months of
age (see Chapter 3.10, Japanese encephalitis).27

Service delivery
General Practitioners, Aboriginal Community Controlled Health Services,
Community Health Services, the Royal Flying Doctor Service and State/Territory
Corrective Services all provide substantial levels of vaccination services to
Aboriginal and Torres Strait Islander people, and are important to the success of
programs to vaccinate Indigenous people. While vaccination coverage estimates

Vaccination for Aboriginal and Torres Strait Islander people 73

2.1Vaccination for
Aboriginal and Torres
Strait Islander people

Studies in far north Queensland and the Kimberley have demonstrated a


favourable impact of the 23-valent pneumococcal polysaccharide vaccine
(23vPPV) on rates of invasive pneumococcal disease in Indigenous adults,18,24,25
but, at a national level, disparities in disease rates between Indigenous and nonIndigenous adults remain. As is the case for influenza and pneumonia, rates of
invasive pneumococcal disease are highest in older Indigenous adults, with rates
around 4 times higher in Indigenous compared to non-Indigenous adults aged
50years.3 Rates in younger adults are slightly lower, but the relative difference
between Indigenous and non-Indigenous is much greater, around 12 times higher
in Indigenous compared to non-Indigenous adults aged 2549years.3 This has
been attributed to a high prevalence of at-risk conditions such as diabetes, renal
disease and excessive alcohol use.26

vary over time and between communities, a relatively consistent finding


has been higher coverage in Aboriginal and Torres Strait Islander people in
remote compared to urban areas.28,29 Recent estimates suggest that, for vaccines
recommended for both Indigenous and non-Indigenous people, coverage is as
high or higher in Indigenous people as non-Indigenous people,3 but vaccination
is more frequently delayed.30 Coverage for vaccines recommended only for
Aboriginal and Torres Strait Islander people is generally lower than for vaccines
which are funded for all people in a particular age group.31 This points to the
importance of identification of Indigenous status, particularly in mainstream
health services, and particularly in urban areas. The use of patient information
systems to record Indigenous status and schedule preventive health services has
the potential to increase opportunistic vaccination and enable the provision of
patient reminders, with improved coverage and timeliness.32

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

74 The Australian Immunisation Handbook 9th Edition

2.2 Vaccination for international travel


Introduction
The number of Australians who travel overseas has increased steadily over
recentyears and now between 3.5 and 4.5 million exits are made annually.
Although many of these trips are to countries where health risks exist, the
majority of Australians travelling overseas do not seek pre-travel health
advice.1 Every year, Australian travellers are injured, become ill, or even die,
while travelling abroad. Some of the infectious diseases that cause some of this
morbidity and mortality are preventable through vaccination.2,3
There is a range of travel vaccines that target infectious diseases that are more
common in different or less-developed environments, and therefore travel
itineraries should be assessed for the level of risk for these diseases.3 Factors
such as the interval between the initial presentation and the departure date,
destination, length of stay, activities during travel, type of accommodation,
personal medical history, age of the traveller, previous vaccination status and
financial constraints, all have a potential impact on vaccine recommendations. It
is important to identify travellers who may be at increased risk for travel related
illness, such as pregnant women, children, people with chronic systemic illness
or people with impaired immunity. Recent immigrants and their Australian-born
children are at particular risk of acquiring some of these infections when they
return to their country-of-origin to visit relatives and friends.4

Common infections acquired by travellers include those which follow ingestion


of contaminated food or water.2,5 Most of these are diarrhoeal diseases due to
enteric pathogens, but infections with extra-intestinal manifestations, such as
hepatitis A and typhoid, are also acquired this way. Vaccines are available for
cholera, hepatitis A and typhoid.
Insect-borne (particularly mosquito) infections, such as malaria and dengue,
are important causes of fever in Australian travellers returning from endemic
areas, southeast Asia and Oceania in particular.5 Japanese encephalitis occurs
throughout much of Asia and probably throughout Papua New Guinea. Yellow
fever occurs only in parts of Africa and Central and South America, while tickborne encephalitis occurs in parts of Europe and Asia. Vaccines are available for
Japanese encephalitis, yellow fever and tick-borne encephalitis.
Vaccine-preventable infections transmitted via respiratory droplets include
influenza, invasive meningococcal disease and measles; influenza may be the most
frequent vaccine-preventable infection among travellers.6 Tuberculosis, although
rare, is mostly acquired by expatriates who live in high-risk areas for long periods.

Vaccination for international travel 75

2.2Vaccination for
international travel

Infections acquired by travellers

Blood-borne infections, such as hepatitis B, hepatitis C and human


immunodeficiency virus (HIV), may pose a threat to some Australian travellers.
In remote areas of some countries, there is the possibility that these viruses
are transmitted by healthcare workers using non-sterile medical equipment.
Hepatitis B vaccine is relevant to many travellers.
Travellers may be exposed to a variety of other exotic infections such as
rabies from dog (and other mammal) bites in many countries, schistosomiasis
after swimming in African lakes, and leptospirosis after rafting or wading in
contaminated streams. Of these, only rabies can be prevented by vaccination.

Practical aspects of travel vaccine administration


Consider each traveller individually, in the context of the specific itinerary. There
is no correct list of vaccines for any single country. Ideally the vaccinations
should be started early, to minimise any adverse events around the time of
departure and allow sufficient time for adequate immunity to develop.
First, consider routine vaccines; all travellers should be up-to-date with
current standard vaccine recommendations. Then consider any other vaccines
that may be relevant to the individuals usual health status, occupation or
lifestyle (eg. pneumococcal polysaccharide vaccine for an elderly person,
hepatitis B vaccine for a first aid officer). These should be offered before
consideration of the travel vaccines.
Travel vaccines should be considered according to risk. Priority should be given
to vaccines for diseases that are common and of significant impact (such as
influenza and hepatitis A), and to those diseases which, although less common,
have severe potential adverse outcomes (such as Japanese encephalitis and
rabies). Booster doses should be considered where appropriate (see Table 2.2.1);
a rapid schedule for a combined hepatitis A/B vaccine is available for those
16years of age with limited time before travel (see the appropriate vaccine
chapters). For children, consider the lower age limits for recommendation of
selected vaccines (see Table 2.2.2).
It is important to document travel vaccines appropriately, not only in the clinics
record but also in a suitable record that can be carried by the traveller.

Vaccines
All intending travellers should have been vaccinated according to the
recommended vaccination schedule for the travellers age. All children should
be vaccinated according to the National Immunisation Program (NIP) schedule.
In exceptional circumstances, the NIP vaccines may be administered at the
minimum age rather than the recommended age (see Section 1.3.5, Catch-up,
Table 1.3.7 Minimum age for the first dose of vaccine in exceptional circumstances).
Children vaccinated using the minimum age rather than the recommended age
may require extra vaccine doses to ensure adequate protection. The minimum

76 The Australian Immunisation Handbook 9th Edition

interval between doses must be adhered to (see Section 1.3.5, Catch-up, Table 1.3.6
Minimum dose intervals for NIP vaccines for children <8years of age).

Measles
Most measles outbreaks now follow infection imported by inadequately
vaccinated young travellers. Therefore, Australians born during or since 1966
who have not received 2 doses of a measles-containing vaccine should be
vaccinated with MMR before travelling. Varicella vaccine should be offered to
travellers who have not had clinical disease or where serology demonstrates
lack of immunity (remembering that 2 doses, separated by at least amonth, are
required by those 14years of age).

Tetanus
Adult travellers should be adequately protected against tetanus before departure,
particularly if there could be delays in accessing health services. They should
receive a booster dose of dT if more than 10years have elapsed since the last
dose. Protection against pertussis may also be offered at this opportunity (as
dTpa) if no previous dose of dTpa has been given.

Poliomyelitis

Influenza and pneumococcal disease


Travellers aged 65years, and those with any medical risk factor, should
receive the seasonal influenza vaccine and should have received the 23-valent
pneumococcal polysaccharide vaccine. All travellers should consider influenza
vaccine, especially when heading to the northern hemisphere winter.

Hepatitis B
All children and adolescents should have been vaccinated against hepatitis B
according to the NIP schedule. As they could be exposed to hepatitis B virus
during unplanned medical procedures, all travellers intending to spend amonth
or more in Central and South America, Africa, Asia or Oceania should be
vaccinated against hepatitis B.

Vaccination for international travel 77

2.2Vaccination for
international travel

All travellers should be age-appropriately immunised against polio. If


travelling to countries where wild polio virus still exists (Afghanistan, India,
Nigeria, and Pakistan), inactivated poliomyelitis vaccine (IPV) should be
offered to those who have not completed a 3-dose primary course of any
polio vaccine, and a single booster dose should be given to those who have
previously completed the primary course. For an up-to-date list of affected
countries see http://www.polioeradication.org.

Hepatitis A
Hepatitis A vaccine should be given to all travellers 1 year of age travelling to
moderately to highly endemic countries (including all developing countries).
There is no place for the routine use of normal human immunoglobulin to
prevent hepatitis A in travellers (see Chapter 3.5, Hepatitis A).

Typhoid
Typhoid vaccine should be given to travellers 2years of age travelling to endemic
regions, which include the Indian subcontinent, most southeast Asian countries,
many south Pacific nations and Papua New Guinea (see Chapter 3.23, Typhoid).

Cholera
Cholera vaccination is rarely indicated for travellers,3 as the risk of acquiring
cholera is extremely low, and the protection is of relatively short duration. It is
only indicated for those travellers at considerable risk, such as those working
in humanitarian disaster situations. However, it can also be considered for
those travellers with achlorhydria and for those at increased risk of severe or
complicated diarrhoeal disease (see Chapter 3.2, Cholera).
Certification of cholera vaccination has been abandoned globally, and no
countries have official entry requirements for cholera vaccination (see Chapter
3.2, Cholera).

Rabies
Travellers to rabies-endemic regions should be advised of the risk, and to avoid
close contact with either wild or domestic animals, and they should be advised
on what to do should they be either bitten or scratched by an animal while
abroad (see Chapter 3.1, Australian bat lyssavirus infection and rabies and also refer
to the World Health Organization website www.who.int).
Pre-travel (ie. pre-exposure) rabies vaccination (or, if appropriate, booster doses)
is recommended for expatriates and travellers who will be spending prolonged
periods (ie. more than amonth) in rabies-endemic areas. (NB. This time interval,
of more than amonth, is arbitrary, and rabies has occurred in travellers following
shorter periods of travel). Vaccination before travel simplifies the management of
a subsequent exposure because fewer doses of vaccine are needed, and because
rabies immunoglobulin (which is often difficult or even impossible to obtain in
many developing countries) is not required.

Japanese encephalitis
Vaccination is recommended for travellers spending amonth or more in either
the rural areas of Asia or in Papua New Guinea, particularly if travel is during
the wet season and/or there is considerable outdoor activity and/or the
standard of accommodation is suboptimal. Vaccination is also recommended for
expatriates spending a year or more in Asia, even if much of the stay is in urban
areas (see Chapter 3.10, Japanese encephalitis).

78 The Australian Immunisation Handbook 9th Edition

Meningococcal infections
All children 12months of age and all teenagers should have received the
meningococcal C conjugate vaccine. In addition, the tetravalent meningococcal
polysaccharide vaccine (4vMenPV) is recommended for those who intend
travelling to parts of the world where epidemics of meningococcal disease occur,
in particular the meningitis belt of sub-Saharan Africa.7 Of note, large epidemics
of meningococcal meningitis occurred in Delhi, India, in 1966, 1985 and 2005.8
The Saudi Arabian authorities require that all pilgrims attending the annual
Hajj have evidence of recent vaccination with 4vMenPV9 (see Chapter 3.12,
Meningococcal disease).

Yellow fever
The World Health Organization no longer routinely reports on yellow fever
infected areas. Rather, the yellow fever vaccine is now recommended for
travellers to yellow fever-endemic countries, in particular those that have
reported yellow fever since 1950 (see Chapter 3.25, Yellow fever, Table 3.25.1 Yellow
fever endemic countries).10
Briefly, provided there is no specific contraindication, the vaccine is
recommended for all those 9months of age travelling anywhere in any country
in West Africa, and for all those 9months of age travelling outside urban areas
of all other yellow fever-endemic countries (see Table 3.25.1).

Tuberculosis

Tick-borne encephalitis
This disease is prevalent in central and northern Europe and across northern Asia
during the summermonths. The vaccine is available only through Special Access
Scheme arrangements inAustralia.

Vaccination for international travel 79

2.2Vaccination for
international travel

Vaccination is generally recommended for tuberculin-negative children <5years


of age who will be living in developing countries for more than 3months.
There is less evidence of the benefit of vaccination in older children and adults,
although consideration should be given to vaccination of tuberculin-negative
children <16years of age who may be living for long periods in high-risk
countries (defined as having an incidence 100 per 100000 population) (see
Chapter 3.22, Tuberculosis).

80 The Australian Immunisation Handbook 9th Edition

JE-VAX

Priorix

Japanese
encephalitis

Measlesmumps-rubella

Live attenuated measlesmumps-rubella viruses

Inactivated Japanese
encephalitis virus

15g haemagglutinin of 2
current influenza A and 1
influenza B strains

10g hepatitis B surface


antigen protein

H-B-VAX II

Various

20g hepatitis B surface


antigen protein

25g S. typhi
polysaccharide and 160 EIA
U inactivated HAV antigen

Engerix-B

Influenza

Hepatitis B

NB. Only
for use in people
16years of age

Vivaxim*

Hepatitis
A/ typhoid
combined

720 EIA U inactivated


HAV antigen and 20g
recombinant hepatitis B
virus surface antigen

50 U inactivated HAV
antigen

VAQTA Adult

Twinrix (720/20)

1440 EIA U inactivated HAV


antigen

Havrix 1440

Hepatitis A/B
combined

160 EIA U inactivated HAV


antigen

Avaxim

Hepatitis A

Main constituents

Brand name

Vaccine
(adults)

0.5mL

1mL

0.5mL

1mL

1mL

1mL
combined
vaccine

1mL

1mL

1mL

0.5mL

Dose
(adults)

IM/SC

SC

IM

IM

IM

IM

IM

IM

IM

IM

Route

Australians born during or since 1966


who do not have documented evidence
of having received 2 doses of a measlescontaining vaccine should receive at
least 1 dose of MMR before travel.

0, 7, 28days

Single dose

0, 1, 6months

0, 1, 6months, or
0, 1, 2, 12months, or

0, 7, 21days, and 12months

Single dose

0, 1, 6months , or

0, 7, 21days, and 12months

0, 6 to 18months

0, 6 to 12months

0, 6 to 12months

Primary schedule

Boosters at 3-yearly intervals.

As different strains circulate from year


to year, annual vaccination with the
current formulation is necessary.

A completed series probably gives


life-long immunity.

A dose of monovalent hepatitis A


vaccine given 636months later
probably gives life-long immunity. The
duration of protection against typhoid
is probably 3years.

A completed series probably gives


life-long immunity to both hepatitis
A and B.

All probably give life-long immunity.

Duration of immunity/booster
recommendations

Table 2.2.1: Dose and routes of administration of commonly used vaccines in adult travellers (15years of age)

Stamaril

Typhim Vi

or

Typherix

Vivotif Oral

Adacel

or

Boostrix

Live attenuated yellow fever


virus

25g purified Vi capsular


polysaccharide

Live attenuated typhoid


bacteria

20 IU tetanus toxoid, 2 IU
diphtheria toxoid, purified
antigens of B. pertussis

20 IU tetanus toxoid, 2 IU
diphtheria toxoid

2.5 IU inactivated rabies


virus antigens

Rabipur
Inactivated Rabies
Vaccine

ADT Booster

2.5 IU inactivated rabies


virus antigens

Single dose

IM/SC

A fourth capsule of oral typhoid vaccine can be given on day 7 (see Chapter 3.23, Typhoid).

10-yearly boosters if at ongoing risk.

Booster doses at 3-yearly intervals

Single dose

IM

Providing pertussis (as well as


tetanus and diphtheria) immunity is
preferred.

Provides protection for 10years.

If at continued high risk of exposure,


either measure rabies antibody
titres (and boost if titres reported as
inadequate) or give single booster
dose 2-yearly.

Repeat 3-dose course after 3years if


3 doses given initially; 4-dose course
after 5years if 4 doses given initially.

0, 7, 28days

0, 7, 28days

Revaccinate 35-yearly if at
continuing risk.

Duration of immunity/booster
recommendations

Days 1, 3 and 5 (+/ day 7)

Oral

IM

IM

IM

IM/SC

Single dose

Primary schedule

This rapid schedule should be used only if there is very limited time before departure to endemic regions.

0.5mL

0.5mL

A single
capsule

0.5mL

0.5mL

1mL

1mL

SC

0.5mL

50g capsular
polysaccharides from N.
meningitidis serogroups A,
C, W135 & Y

Mrieux
Inactivated Rabies
Vaccine

Menomune

Route

Dose
(adults)

Main constituents

* Vivaxim is registered for use in people aged 16years.

Yellow fever

Typhoid

+ pertussis
(dTpa)

Tetanus,
diphtheria (dT)

Rabies
(pre-exposure
prophylaxis)

Mencevax ACWY

Meningococcal
(tetravalent
polysaccharide)

or

Brand name

Vaccine
(adults)

2.2Vaccination for
international travel

Vaccination for international travel 81

82 The Australian Immunisation Handbook 9th Edition

Rabipur

Mrieux

Rabies

Mencevax ACWY
or Menomune

Meningococcal ACW135Y

JE-VAX

No lower age limit

2years

1 year

1 year

Twinrix (720/20)

Japanese encephalitis

1 year

Twinrix Junior (360/10)

1.0mL IM

1.0mL IM/SC

0, 7, 28days

0, 7, 28days

Pre-exposure:

Single dose

0, 7, 28days

>3years of age:
1.0mL SC

0.5mL SC

0, 7, 28days

*0, 6 to 12months

13years of age:
0.5mL SC

1.0mL IM

0, 1, 6months

0, 6 to 18months

0.5mL IM

1 year

VAQTA Paediatric/ Adolescent

0.5mL IM

0, 6 to 12months

0.5mL IM

Hepatitis A/B combined

0, 6 to 12months

0.5mL IM

2years

Primary schedule

2years

Dose/route

Havrix Junior

Lower age limit

Avaxim

Hepatitis A

Vaccine

Table 2.2.2: Recommended lower age limits of travel vaccines for children

The doses of rabies vaccines for pre-exposure are the same


for both children and adults (1.0mL).

Should be preceded by MenCCV by at least 2weeks.

Revaccinate 35-yearly if at continuing risk.

Recommended for travellers spending more than 4weeks in


rural areas of Asia and Papua New Guinea, or those staying
in urban areas of Asia for more than 1 year.

Recommended for travel to developing countries.

Recommended for travel to developing countries.

Comments

See Chapter 2.3, Groups with special vaccination requirements and the specific vaccine chapters for recommendations for travellers
who are either pregnant or have impaired immunity. Children should receive the relevant travel vaccines, according to age (see
Table 2.2.2). Particular effort should be made to encourage the families of recent migrants to Australia to seek health advice before
travelling to their country of origin to visit relatives and friends.11

Vaccinating the traveller with special risk factors

0.5mL IM/SC

0.5mL IM

Oral capsule

Dose/route

Single dose

Single dose

One capsule ondays 1, 3,


and 5 (+/ day 7)

Primary schedule

Yellow fever vaccine is contraindicated in infants <9months


of age.

Do not give live oral vaccine with antibiotics or antimalarials. Do not give within 8 hours of inactivated oral
cholera vaccine.

Recommended for travel to developing countries.

Comments

Full reference list available on the electronic Handbook or website http://immunise.health.gov.au.

References

It should be noted that information on travellers risks is changing constantly. Up-to-date knowledge requires an understanding
of the changing epidemiology of a variety of infectious and emerging diseases. The World Health Organizations comprehensive
publication International Travel and Health is available at www.who.int/ith and the CDCs publication Health Information for
International Travel, 20052006 (the Yellow Book) is available at www.cdc.gov/travel/index.htm. As recommendations for specific
countries change frequently, such sources should be checked regularly.

Further information

A fourth capsule of oral typhoid vaccine can be given on day 7 (see Chapter 3.23, Typhoid).

* This schedule is not recommended if prompt protection against hepatitis B is required.

Stamaril

9months

2years

Typherix or Typhim Vi (parenteral


vaccine)

Yellow fever

6years

Lower age limit

Vivotif Oral
(oral live vaccine)

Typhoid

Vaccine

2.2Vaccination for
international travel

Vaccination for international travel 83

2.3 Groups with special


vaccination requirements
This chapter considers the use of vaccines in people who have special vaccination
requirements, those who may experience more frequent adverse events, and
those who may have a suboptimal response to vaccination. Recommendations for
vaccination of those at occupational risk are also included.

2.3.1 Vaccination of children who have had a serious


adverse event following immunisation (AEFI)
Children who have had a serious AEFI (other than a contraindication, such as
anaphylaxis) may be subsequently vaccinated under close medical supervision
(see Appendix 6, Definitions of adverse events following immunisation). Not all States
and Territories offer an adverse event immunisation clinic. However, in States or
Territories where there are no clinics, there is often a paediatrician or infectious
diseases specialist who will review families who have concerns regarding future
vaccinations following a previous adverse event.
To make an enquiry, or for more information about making a referral to a
vaccination serious adverse events service, please go to the website below:
http://immunise.health.gov.au/internet/immunise/publishing.nsf/Content/
provider-reactions.

2.3.2 Vaccination of women planning pregnancy,


pregnant or breastfeeding women, and preterm infants
(i) Women planning pregnancy
The need for measles, mumps, rubella, varicella, diphtheria, tetanus and
pertussis vaccination should be assessed as part of any pre-conception health
check. Where previous vaccination history or infection is uncertain, relevant
serological testing should be undertaken to ascertain immunity. Influenza vaccine
is recommended routinely and pneumococcal vaccination is recommended
for women with risk factors, including smokers. Women receiving live viral
vaccines must be advised against falling pregnant within 28days of vaccination.
Please refer to individual disease chapters for more information about primary
vaccination for these diseases.

84 The Australian Immunisation Handbook 9th Edition

(ii) Pregnancy
Although the use of most vaccines during pregnancy is not usually
recommended on precautionary grounds, there is no convincing evidence that
pregnancy should be an absolute contraindication to the use of any vaccine,
particularly inactivated vaccines. The only exception is vaccinia virus (smallpox
vaccination), which has been shown to cause fetal malformation. There is some
evidence, however, that fever per se is teratogenic.1,2 With the exception of the use
of influenza vaccine, the NHMRC takes the conservative position that the use of
vaccines during pregnancy might cause fever and should be avoided other than
in situations of increased risk, where the benefits of protection from vaccination
outweigh the risks. Eliminating the risk of exposure to vaccine-preventable
diseases during pregnancy (eg. by changing travel plans, avoiding high-risk
behaviours or occupational exposures) is an alternative to vaccination.
Live attenuated vaccines are contraindicated for pregnant women because of
the hypothetical risk of harm to the fetus should transmission occur. If a live
attenuated vaccine is inadvertently given to a pregnant woman, or if a woman
becomes pregnant within 4weeks of vaccination, she should be counselled
about the potential transmission, albeit extremely unlikely, to the fetus. There is,
however, no indication to consider termination of a pregnancy in this situation.
The following table may be used as a guide to the recommended use of
vaccinations in pregnancy. Information regarding pregnancy status in women of
reproductive age should be part of the routine pre-vaccination screening checklist
(see Section 1.3.4, Pre-vaccination screening).

2.3 Groups with special


vaccination requirements

Groups with special vaccination requirements 85

86 The Australian Immunisation Handbook 9th Edition


Hypothetical risk only. Despite concerns that attenuated rubella vaccine virus might cause
congenital abnormalities, rubella vaccine (either monovalent or as MMR) has been given to
pregnant women (usually inadvertently) without harm to the fetus. Even though the rubella
vaccine virus can infect the fetus if given in early pregnancy, there is no evidence that it causes
congenital rubella syndrome in infants born to susceptible mothers vaccinated during pregnancy
and, in particular, rubella vaccination during pregnancy is not an indication for termination.3

Should not be given to women who are pregnant or considering becoming pregnant.
Pregnancy should be avoided for 3months after vaccination.
Hypothetical risk only. Congenital varicella syndrome has (to date) not been identified in
women who have been inadvertently vaccinated in early pregnancy.4 This provides some
reassurance of the safety of the vaccine.

Contraindicated.

Contraindicated.

Contraindicated.

Contraindicated, unless travelling to yellow fever


endemic area.

Measles-mumpsrubella (MMR) vaccine

Smallpox vaccine

Varicella vaccine

Yellow fever vaccine

The administration of yellow fever vaccine in early pregnancy is not an indication for termination.

Hypothetical risk only. Yellow fever vaccine has been given to a large number of pregnant
women with no adverse outcomes.5 Pregnant women who travel to a yellow fever-endemic
area against medical advice should receive yellow fever vaccine.

Women of child-bearing age should avoid becoming pregnant for 28days after vaccination.

It is standard practice to test all pregnant women for immunity to rubella, and to vaccinate
susceptible women as soon as possible after delivery (preferably using MMR).

Women of child-bearing age should avoid pregnancy for 28days after vaccination.

Comments

Recommendation

Viral

Live attenuated vaccines

Contraindicated.

Studies in animals are inadequate but available data show no evidence of an increased
occurrence of fetal damage with oral live attenuated vaccine. Inactivated typhoid Vi
polysaccharide vaccine is preferred.

Rotavirus vaccine can be safely administered to household contacts of pregnant women.

Contraindicated.

Rotavirus vaccine

Oral typhoid vaccine

Hypothetical risk only. BCG has not been shown to cause fetal damage.

Contraindicated.

BCG vaccine
(Live attenuated strain
M. bovis)

Not registered for use in adults.

Comments

Recommendation

Bacterial

Live attenuated vaccines

Table 2.3.1: Vaccinations in pregnancy

Comments

Inadequate information on safety of oral cholera vaccine in pregnancy.


Data on use of adolescent/adult formulation dTpa during pregnancy are not available, so it
should be given in pregnancy only when the possible advantages outweigh the possible risks
to the fetus. All women who are planning pregnancy should be encouraged to receive a single
dose of dTpa before pregnancy; if not given before pregnancy, it should be given as soon as
possible after delivery.
Available clinical data suggest that it is unlikely that use of Hib vaccine in pregnant women
would have any deleterious effects on the pregnancy.
Although no clinical study data are available on the use of MenCCV in pregnant women, it is
unlikely that it would have any deleterious effects on the pregnancy.

No adverse effects when administered in pregnancy. Data are limited to clinical trials and
deferral of vaccine is recommended unless there is an increased risk of IPD. Women of
reproductive age with known risk factors for IPD (including smokers) should be vaccinated
before planned pregnancy.

Safety of use in pregnancy has not been established.


There is no evidence of risk to the fetus from vaccination with Vi polysaccharide vaccine.

Not recommended.

Recommended for pregnant women who work


in close contact with infants eg. childcare,
neonatal units.

Recommended for pregnant women at increased


risk of Hib disease (eg. hyposplenia, asplenia).

Recommended for pregnant women at increased


risk of meningococcal disease (eg. hyposplenia,
asplenia), or possible exposure to serogroup C.

Recommended for pregnant women at increased No documented adverse events in either pregnant women or their newborns when vaccinated
risk of meningococcal disease who have not been with 4vMenPV administered in the second and third trimesters of pregnancy. The number of
vaccinated with 4vMenPV in the past 3years (eg. pregnant vaccinees reported in the literature is small.
hyposplenia, asplenia), or possible exposure to
serogroup A, W135 or Y.
Vaccination during pregnancy has not been evaluated for potential harmful effects to mother or
fetus. Although unlikely to result in adverse effects, the vaccine is currently only registered for
use in children 9years of age.

Recommendation

Not recommended.

Recommended for pregnant women at increased


risk of invasive pneumococcal disease (IPD) (eg.
asplenia, impaired immunity, chronic illness, CSF
leak) who have not received 23vPPV in the past
5years (and provided they have not received 2
previous doses).

Not recommended.

In pregnant women travelling to endemic


countries where water quality and sanitation
is poor.

Bacterial

Cholera (oral) vaccine

Adolescent/adult
formulation dTpa
vaccine

Haemophilus influenzae
type b (Hib) vaccine

Meningococcal C
conjugate vaccine
(MenCCV)

Meningococcal
polysaccharide vaccine
(4vMenPV)

7-valent pneumococcal
conjugate vaccine
(7vPCV)

23-valent
pneumococcal
polysaccharide vaccine
(23vPPV)

Q fever vaccine

Typhoid Vi
polysaccharide vaccine

Inactivated vaccines

2.3 Groups with special


vaccination requirements

Groups with special vaccination requirements 87

88 The Australian Immunisation Handbook 9th Edition


Hepatitis A vaccine should only be given to pregnant women who are non-immune and where
there is a clear indication. As for any inactivated viral vaccines, although data are limited, no
adverse effects on the developing fetus are expected.

Hepatitis B vaccine should only be given to pregnant women who are non-immune and where
there is a clear indication. As for any inactivated viral vaccines, although data are limited, no
adverse effects on the developing fetus are expected.
There are no concerns that HPV vaccines are teratogenic and animal studies have found no
evidence of teratogenicity or adverse fetal outcomes. However, where vaccine has inadvertently
been administered during pregnancy, further doses should be deferred until after delivery.
There is no evidence of congenital defects or adverse effects on the fetus of women who are
vaccinated against influenza in pregnancy.

No adverse effects on pregnancy have been attributed to JE vaccine, whereas JE infection is


associated with miscarriage.
IPV should only be given to pregnant women when clearly indicated. There is no convincing
evidence of risk to the fetus from IPV administered in pregnancy.
Pregnancy is never a contraindication to rabies vaccination in situations where there is a
significant risk of exposure (related to occupation or travel), or where there has been a possible
exposure to rabies virus or Australian bat lyssavirus.

Recommended for susceptible pregnant


women travelling to areas of moderate to high
endemicity or who are at increased risk of
exposure through lifestyle factors, or where
severe outcomes may be expected (eg. preexisting liver disease).

Recommended for susceptible pregnant women


for whom this vaccine would otherwise be
recommended.

Not recommended.

Recommended for all pregnant women who


will be in the second or third trimester during
the influenza season, including those in the first
trimester at the time of vaccination.

Recommended for pregnant women at risk of


acquiring JE.

Recommended for pregnant women at risk


of poliovirus exposure (eg. travel to endemic
countries).

Recommended for pregnant women for whom


this vaccine would otherwise be recommended
(eg. travellers to rabies endemic countries).

Hepatitis A vaccine

Hepatitis B vaccine

Human papillomavirus
(HPV) vaccine

Influenza vaccine

Japanese encephalitis
(JE) vaccine

Inactivated polio
vaccine (IPV)

Rabies vaccine

Toxoids are safe in pregnancy.


There is no known risk to the fetus from passive immunisation of pregnant women with
immunoglobulins.

Recommended for pregnant women.

Recommended for susceptible pregnant women


exposed to: measles, hepatitis A, hepatitis B,
rabies or Australian bat lyssavirus, varicella
viruses and tetanus.

Tetanus/diphtheria
toxoid

Pooled or
hyperimmune
immunoglobulins

Toxoids and immunoglobulins

Comments

Recommendation

Viral

Inactivated vaccines

Contact between pregnant women and individuals


who have recently received live vaccines
Although there is no risk of transmission of the MMR vaccine viruses (MMR
vaccine viruses are not transmissible), and an almost negligible risk of
transmission of varicella vaccine virus, there is a very small risk of transmission
of the rotavirus vaccine viruses to a susceptible pregnant woman. However, there
is no evidence that there is any risk to the fetus if pregnant women are in contact
with recently vaccinated individuals. Therefore, it is safe to administer varicella
vaccine and rotavirus vaccine to household contacts of pregnant women.

(iii) Breastfeeding and vaccination


The rubella vaccine virus may be secreted in human breast milk and transmitted
to breastfed infants but, where infection has occurred in an infant, it has been
mild. Otherwise, there is no evidence of risk to the breastfeeding baby if the
mother is vaccinated with any of the live or inactivated vaccines described in
this Handbook. Breastfeeding does not adversely affect immunisation and is not a
contraindication for the administration of any vaccine to the baby.

(iv) Preterm babies


Preterm (premature) infants have a special need for protection and, despite their
immunological immaturity, they generally respond well to vaccines. Provided they
are well and there are no contraindications to vaccination they should be vaccinated
according to the recommended schedule at the usual chronological age.6-14
Routine childhood vaccines can cause an increase in apnoea in preterm babies
vaccinated in hospital, particularly babies still requiring special care, but these
are generally self-limiting and do not affect the clinical course.8 Preterm babies in
hospital should be monitored for apnoea or bradycardia for up to 48 hours post
vaccination.6-8 Vaccinations have not caused an increase in apnoeas in babies at
home, and are not associated with an increased risk of SIDS.6-8
Vaccines as recommended on the National Immunisation Program (NIP)
schedule

Pneumococcal vaccines
All preterm babies born at less than 28weeks gestation or with chronic lung
disease should be offered the 7-valent pneumococcal conjugate vaccine at 2, 4
and 6months of age, with a fourth dose at 12months of age, and a 23-valent
pneumococcal polysaccharide vaccine booster at 45years of age (see Chapter
3.15, Pneumococcal disease).

Groups with special vaccination requirements 89

2.3 Groups with special


vaccination requirements

Preterm babies produce good antibody responses to most vaccines in the NIP.
They should be vaccinated at the standard recommended ages without correction
for prematurity.14

Haemophilus influenzae type b vaccine


Some smaller preterm babies do not respond as well as term babies to PRP-OMP
(Liquid PedvaxHIB or COMVAX) Hib vaccine.9-14 When PRP-OMP is used in an
extremely preterm baby (<28weeks gestation or <1500 g birth weight), an extra
dose of vaccine should be given at 6months of age (ie. doses should be given at
2, 4, 6 and 12months of age) (see Chapter 3.4, Haemophilus influenzae type b).
Hepatitis B vaccine
Preterm babies do not respond as well to hepatitis B-containing vaccines as
term babies.7,12,13,15 Thus, for babies born at <32weeks gestation or <2000 g birth
weight, it is recommended to give vaccine at 0, 2, 4 and 6months of age and
either:
(a) measure anti-HBs at 7months of age and give a booster at 12months of age if
antibody titre is <10mIU/mL, or
(b) give a booster at 12months of age without measuring the antibody titre.
(See Chapter 3.6, Hepatitis B.)
Influenza vaccine
Preterm infants with ongoing problems at 6months of age, particularly
respiratory, cardiac or neurological disease, should receive influenza vaccine.

2.3.3 Vaccination of individuals with impaired


immunity due to disease or treatment16-18
The vaccination of individuals with impaired immune systems presents several
problems. First, the immune response to vaccines may be inadequate and,
second, there is a risk that some live vaccines may themselves cause progressive
infection. Degrees of impaired immunity vary from insignificant to profound,
and this should be taken into account when considering a vaccination schedule,
as should the risk of acquiring the vaccine-preventable diseases.
Although it may seem logical to give higher or more frequent doses of vaccines
to these patients, in many cases there are insufficient data to advocate such
measures. Because of the uncertainty of the immune response in some patients
with impaired immunity, it may be useful to measure post-vaccination antibody
titres in groups such as children who have received haematopoietic stem cell
transplants (see 2.3.3.3 Re-vaccination following haematopoietic stem cell
transplantation (HSCT) below).
Administration of certain vaccines is a priority for some patients with medical
conditions that increase the risk from infectious diseases, even in the absence
of specific immune defects. These include: the use of influenza vaccine in
individuals with severe asthma, chronic lung disease, congenital heart disease,
diabetes and Down syndrome; pneumococcal conjugate vaccine in children

90 The Australian Immunisation Handbook 9th Edition

with renal failure, persistent nephrotic syndrome and certain anatomical


abnormalities; and pneumococcal polysaccharide vaccine in adults with
certain chronic medical conditions. See the appropriate chapters for current
recommendations.

Live viral and bacterial vaccines


Although most live vaccines are contraindicated in patients with significantly
impaired immunity, the risk of progressive infection varies. The following is a list
of current recommendations:
Vaccines for smallpox (vaccinia virus) and tuberculosis (BCG) are always
contraindicated.
Live vaccines such as MMR and varicella vaccines must not be given to
people with severely impaired immunity, but are safe to be given to the
siblings or other household contacts of such people.
MMR and varicella vaccines may be given to children with HIV infection
with mildly impaired immunity (see 2.3.3.4 HIV-infected individuals
below).
Travellers with impaired immunity should not receive oral typhoid vaccines.
Use parenteral typhoid Vi polysaccharide vaccine instead.
Yellow fever vaccine is contraindicated in travellers with impaired
immunity going to endemic countries. If they must proceed with the travel,
they should obtain a letter from a doctor, clearly stating the reason for
withholding the vaccine. The letter should be formal, signed and dated, and
on the practices letterhead.

Household contacts vaccinated with live vaccines who


live with a person who has impaired immunity

Groups with special vaccination requirements 91

2.3 Groups with special


vaccination requirements

Healthy siblings and household contacts of children with impaired immunity


should be vaccinated with MMR, varicella and rotavirus vaccines (where
indicated) to prevent them from infecting the children with impaired
immunity. Although there is no risk of transmission of the MMR vaccine
viruses, and an almost negligible risk of transmission of varicella vaccine
virus, there is a small risk of transmission of the rotavirus vaccine viruses
(see Chapter 3.18, Rotavirus and Chapter 3.24, Varicella). Annual influenza
vaccination is recommended for contacts (including children 6months of age)
of people with impaired immunity.

Influenza and pneumococcal vaccines


Morbidity and mortality from influenza and invasive pneumococcal disease
are increased in all people with severely impaired immunity. Annual influenza
vaccination should be given to all people 6months of age with severely
impaired immunity. Such individuals should also receive either the 7-valent
pneumococcal conjugate vaccine (7vPCV), or 23-valent pneumococcal
polysaccharide vaccine (23vPPV), depending on their age (see Chapter 3.15,
Pneumococcal disease). Although the immune response to 23vPPV may be
suboptimal in those who most need protection, the vaccine is nevertheless
strongly recommended for these individuals.
While it may seem logical to give 7vPCV followed by 23vPPV to adults with
impaired immunity, studies evaluating the effectiveness of such a regimen are
not yet available.

Impaired immunity associated with corticosteroid administration


In adults, daily doses of oral corticosteroids in excess of 60mg of prednisolone
(or equivalent) and, in children, doses in excess of either 2mg/kg per day for
more than aweek or 1mg/kg per day for more than 4weeks, are associated with
significantly impaired immunity. However, even lower doses may be associated
with some impairment of the immune response.19
Children on daily doses of 2mg/kg per day of systemic corticosteroids for
less than 1week, and those on lower doses of 1mg/kg per day or alternate-day
regimens for periods of up to 4weeks, may be given live viral vaccines.
Children receiving >2mg/kg per day or 20mg per day in total of prednisolone
(or equivalent) for >14days can receive live viral vaccines after corticosteroid
therapy has been discontinued for at least 1month.
For adults treated with systemic corticosteroids in excess of 60mg per day, live
vaccines (such as MMR and varicella vaccines) should be postponed until at least
3months after treatment has stopped.

2.3.3.1 Oncology patients16,20-24


Paediatric and adult patients undergoing cancer chemotherapy who have
not completed a primary vaccination schedule before diagnosis
Live viral vaccines, including varicella and MMR vaccines, are contraindicated
in cancer patients receiving immunosuppressive therapy and/or with
poorly controlled malignant disease. These vaccines may be administered
to seronegative children at least 3months after completion of chemotherapy
and/or high-dose steroid therapy, provided the underlying malignancy is in
remission. Administration of live viral vaccines (MMR, varicella or MMRV
[when available]) should be deferred if blood products or immunoglobulins have
been recently administered (see Table 2.3.5 Recommended intervals between either
immunoglobulins or blood products and MMR, MMRV or varicella vaccination).

92 The Australian Immunisation Handbook 9th Edition

Influenza vaccination is recommended annually in all cancer patients 6months


of age and should be started as close to the time of cancer diagnosis as possible.
During chemotherapy, and for 6months afterwards, patients may receive
inactivated vaccines (eg. DTPa if <8years old, Hib if <5years old, hepatitis B,
IPV) according to the routine vaccination schedule, but it should be remembered
that patients are unlikely to mount a full immune response while they are on
therapy. Antibody responses to hepatitis B should be checked 4weeks after
completing the third dose and, where antibody titres are <10 IU/mL, HBsAg
carriage should be investigated. If HBsAg negative, then patients should be
given a fourth double dose of vaccine or a further 3 doses of vaccine atmonthly
intervals (see Chapter 3.6, Hepatitis B).
Vaccines should not be administered during times of severe neutropenia
(absolute neutrophil count <0.5 x 109/L), to avoid precipitating an acute
febrile episode.
Pneumococcal vaccination is recommended in oncology patients with an increased
risk of invasive pneumococcal disease (IPD), especially patients with underlying
haematological malignancies (multiple myeloma, Hodgkins lymphoma, nonHodgkins lymphoma, chronic lymphocytic leukaemia). In patients 10years
of age, 23vPPV should ideally be given as early as possible after diagnosis and
before chemotherapy and/or radiotherapy is initiated.25,26 When this is not
practicable, vaccination should be given after completion of chemotherapy.27 For
children <10years of age with haematological malignancies, primary and catchup pneumococcal vaccination should be administered as detailed in Table 1.3.11
Recommendations for pneumococcal catch-up vaccination for children 5years of age with
underlying medical conditions (see footnote accompanying this Table).
Any deviations from these guidelines should be discussed with an oncologist.
Paediatric and adult patients with cancer who have completed cancer
therapy and have received a primary course of vaccination before diagnosis

DTPa if <8years of age (use dT or adolescent/adult formulation dTpa if


8years of age),
MMR, IPV, hepatitis B, 7vPCV and Hib (if <5years of age or with previous
splenectomy/hyposplenism).
These vaccines may be given without checking antibody titres beforehand, and
may be given together on 1 day. Measles and rubella antibody status should be
checked 6 to 8weeks after vaccination. Patients who have not seroconverted
should receive a further dose.

Groups with special vaccination requirements 93

2.3 Groups with special


vaccination requirements

The following schedule of booster vaccination is recommended if the patient


is well and infection-free 6months after chemotherapy, and if the underlying
disease is in remission:

Children who are seronegative to varicella-zoster virus, especially those with


acute lymphoblastic leukaemia, should receive a 2-dose schedule of varicella
vaccine, at least 3months after chemotherapy has been ceased.28 Administration
of live vaccines (MMR, varicella or MMRV [when available]) should be deferred
if blood products or immunoglobulins have been recently administered (see
Table 2.3.5 Recommended intervals between either immunoglobulins or blood products
and MMR, MMRV or varicella vaccination).

2.3.3.2 Solid organ transplant recipients23


For solid organ transplant (SOT) recipients, depending on the transplanted
organ, and to prevent rejection, differing doses of immunosuppressive agents
are needed, which may influence the effectiveness of vaccines. Where possible,
children undergoing solid organ transplantation should be vaccinated well
before transplantation, and inactivated vaccines can be used 6 to 12months after
transplantation. Live vaccines are contraindicated in most post-transplantation
protocols due to concerns of disseminated infection, although data in this
population are limited. Recommended vaccinations for child and adult SOT
recipients are given in Table 2.3.2.

94 The Australian Immunisation Handbook 9th Edition

Yes

Yes

Hepatitis A vaccine

Hepatitis B vaccine

[If <10years
of age see
Table 1.3.11.]

Yes

7-valent pneumococcal Yes, if <10years


of age
conjugate vaccine
(7vPCV)

23-valent
pneumococcal
polysaccharide vaccine
(23vPPV)

Yes, depending on
serological status

Yes, if seronegative

Adult

Yes

Yes

Yes

Child

Yes, depending on
serological status

Yes, if seronegative

Adult

Vaccines recommended after


transplantation if not given
beforehand

Yes

[If <10years
of age see
Table 1.3.11.]

Yes

Yes, if <10years
of age

Yes

Annual vaccination starting before transplantation for people 6months of age.

Yes

Hib vaccine

Influenza vaccine

Child

Vaccine

Vaccines recommended before


transplantation

See Table 3.15.3 Revaccination with


23vPPV for people 10years of age.

For children <10years of age, 7vPCV


should be administered as detailed
in Table 1.3.11 Recommendations for
pneumococcal catch-up vaccination
for children 5years of age with
underlying medical conditions (see
footnote accompanying this Table).

The primary schedule should be


completed before transplantation.

Accelerated schedules can be


used (see Table 3.6.2 Accelerated
hepatitis B vaccination schedules).

Recommended for all


seronegative SOT recipients.

Recommended for all


seronegative SOT recipients.

If possible complete vaccination at least


6weeks before transplantation.23

Comments

Table 2.3.2: Recommendations for vaccinations for solid organ transplant (SOT) recipients

2.3 Groups with special


vaccination requirements

Groups with special vaccination requirements 95

96 The Australian Immunisation Handbook 9th Edition

Yes

Diphtheria-tetanuspertussis vaccine

Yes,
unless 2 previous
documented doses

Yes

Yes

MMR vaccine

Varicella vaccine
Yes

Yes

Yes

Yes, provided
dTpa has not been
given previously

Yes, if no booster
in past 10years

Adult

Meningococcal
Yes, if >2years
polysaccharide vaccine of age
(4vMenPV)

Meningococcal C
conjugate vaccine
(MenCCV)

Yes, if 1
year of age

Yes

Inactivated poliovirus
vaccine (IPV)

(DTPa for children


<8years of age; dTpa
for people 8years of
age)

Child

Vaccine

Vaccines recommended before


transplantation

Contraindicated

Contraindicated

Yes, if >2years
of age

Yes, if 1
year of age

Yes

Yes

Child

Yes

Yes

Yes, provided
dTpa has not been
given previously

Yes, if no booster
in past 10years

Adult

Vaccines recommended after


transplantation if not given
beforehand

Vaccination should be completed before


transplantation provided the recipient is
no longer on immunosuppressive therapy.

The primary schedule should be


completed before transplantation
provided the recipient is no longer
on immunosuppressive therapy.

Give 4vMenPV at an interval of at


least 2weeks after MenCCV.

The primary schedule should be


completed before transplantation.

The primary schedule should be


completed before transplantation.

Comments

2.3.3.3 Re-vaccination following haematopoietic


stem cell transplantation (HSCT)29-33
Haematopoietic stem cells are sourced from peripheral blood, bone marrow or
umbilical cord. Protective immunity to vaccine-preventable diseases is partially or
completely lost following either allogeneic or autologous stem cell transplantation.
Impaired immunity following allogeneic transplantation is caused by a combination
of the preparative chemotherapy given before transplantation, graft-versus-host
disease (GVHD), and immunosuppressive therapy following transplantation.
Persisting impaired immunity is common, particularly in patients with chronic
GVHD. Immunity is also impaired in autologous stem cell transplant recipients due
to high-dose chemotherapy and radiotherapy, but GVHD is not a concern as donor
and recipient are the same. In most cases, autologous transplant recipients will
recover their immunity more quickly than allogeneic transplant recipients.
Separate transplant schedules for autologous and allogeneic transplant recipients
have not been supported in published guidelines because of limited data. For
practical purposes, a similar schedule is therefore recommended, regardless of
donor source (peripheral blood, bone marrow or umbilical cord), preparative
chemotherapy (ablative or reduced intensity), or transplant type (allogeneic or
autologous).32,34
HSCT recipients with ongoing GVHD or remaining on immunosuppressive
therapy should not be given live vaccines. Chronic GVHD (cGVHD) is associated
with functional hyposplenism and patients are therefore susceptible to infections
with encapsulated organisms, especially Streptococcus pneumoniae. For patients
with cGVHD who remain on active immunosuppression, antibiotic prophylaxis
is recommended.35
The immune response to vaccinations is usually poor during the first
6months after HSCT. Donor immunisation with hepatitis B, tetanus, Hib and
pneumococcal conjugate vaccines before stem cell harvesting has been shown to
elicit improved early antibody responses in HSCT recipients vaccinated in the
post-transplantation period.36-39 However, practical and ethical considerations
currently limit the use of donor immunisation.

A recommended schedule of vaccination is outlined in Table 2.3.3.31,32,34

Groups with special vaccination requirements 97

2.3 Groups with special


vaccination requirements

Routine serological testing for several infectious agents increases costs,


and antibody levels conferring protective immunity are poorly defined.
For those vaccines that are recommended for all HSCT recipients (tetanus,
diphtheria, polio, influenza, pneumococcal, Hib), pre-vaccination testing is not
recommended as the response to a primary course of these vaccines is generally
adequate. The serological response to pneumococcal polysaccharide vaccine is
less predictable. Pneumococcal serology is only available in a few specialised
laboratories and is not routinely recommended. Immunity testing before and
after vaccination for hepatitis B, measles, rubella and varicella is recommended,
as antibody levels will determine the need for revaccination.34

Table 2.3.3: Post-transplantation vaccination schedules for allogeneic and


autologous haematopoietic stem cell transplant recipients32,34
Vaccine

Months after HSCT

Comments

12

14

24

Yes

Yes

Yes

Hib

Yes

Yes

Yes

Hepatitis A

Not routinely recommended, see Chapter 3.5, Hepatitis A.

Hepatitis B

Yes

Influenza

Annual vaccination for life, starting 6months post HSCT, for people 6months
of age.

MMR

No

MenCCV

Yes, 1
year of
age

People 1 year of age should receive 1


dose of MenCCV.

4vMenPV

Yes,
2years
of age (see
comment)

People 1 year of age should receive 1


dose of MenCCV (as above). This should
be followed by a dose of 4vMenPV
when 2years of age or, if already aged
>2years, give after an interval of at least
2weeks following the MenCCV.

7vPCV

Although there are limited data on the effectiveness of 7vPCV in HSCT recipients,
vaccination is recommended for children 9years of age starting 6months post
HSCT (see Table 3.15.1 Summary table pneumococcal vaccination schedule for
children 9years of age).

23vPPV

Yes

Diphtheria-tetanuspertussis

(DTPa for children


<8years of age; dTpa for
people 8years of age)

Yes

No

Yes

Yes

For recipients 8years of age, give first


dose as dTpa followed by 2 doses dT. If
dT unavailable, dTpa may be used for all
3 doses.

High dose (H-B-VAX II dialysis


formulation) vaccine is recommended.

Vaccination of measles or rubella


seronegative HSCT recipients at
24months post HSCT is recommended,
provided that immunosuppressive
therapy has been discontinued, there is
no chronic GVHD, and cell-mediated
immunity has been reconstituted.

See Table 3.15.3 Revaccination with


23vPPV for people 10years of age.
Adjunctive antibiotic prophylaxis is
recommended for patients with chronic
GVHD.

IPV

Yes

Yes

Yes

Varicella vaccine

No

No

Yes

98 The Australian Immunisation Handbook 9th Edition

Vaccination of seronegative HSCT


recipients at 24months post HSCT
is recommended, provided that
immunosuppressive therapy has been
discontinued, there is no chronic GVHD,
and cell-mediated immunity has been
reconstituted.

2.3.3.4 HIV-infected individuals


Vaccination schedules for HIV-infected patients should be determined by the
patients age, degree of impaired immunity (CD4 count) and the risk of infection.
Children with perinatally acquired HIV differ substantially from adults as
immunisation and first exposure to vaccine antigens occurs after HIV infection,
whereas for adults, most vaccines are inducing a secondary immune response.40
HIV-infected individuals of any age who are well controlled on combination
antiretroviral therapy (undetected or low viral load with good preservation of
CD4 lymphocyte count) are likely to respond well to vaccines.
HIV-infected patients should be vaccinated as follows:
Diphtheria-tetanus-pertussis (DTPa/dTpa), Hib and IPV vaccines use the
standard schedule.41
MMR vaccine should be routinely administered to HIV-infected children
at 12months of age unless they have severely impaired immunity. Table
2.3.4 shows age-specific definitions of moderately and severely impaired
immunity. Measles may cause severe disease in HIV-infected children and
children with severely impaired immunity who are exposed to measles
should, therefore, be given normal immunoglobulin (in a dose of 0.5mL/kg),
regardless of their vaccination status.40
While varicella vaccine is contraindicated in adults with HIV, its use may
be considered for asymptomatic or mildly affected children 12months and
<13years of age.42 The Advisory Committee on Immunization Practices
(ACIP) recommends use of the vaccine, given in 2 doses, 3months apart, in
children with age-specific CD4 T-lymphocyte percentages greater than 25%.43
Table 2.3.4: Immunological categories based on age-specific CD4 counts and
percentage of total lymphocytes44
Category

<12months

15years

6years

CD4 per
L

CD4 per
L

No evidence
of impaired
immunity

1500

25

1000

25

500

25

Moderately
impaired
immunity

7501499

1524

500999

1524

200499

1524

Severely
impaired
immunity

<750

<15

<500

<15

<200

<15

Groups with special vaccination requirements 99

2.3 Groups with special


vaccination requirements

CD4 per
L

Pneumococcal disease, both respiratory and invasive, is a frequent cause of


morbidity in HIV-infected children and adults. Infants and children <10years
of age should be vaccinated with the 7vPCV (see Table 3.15.1 Summary table
pneumococcal vaccination schedule for children 9years of age) and older children
and adults should be vaccinated with the 23vPPV (see also Chapter 3.15,
Pneumococcal disease).45,46
Influenza vaccine is recommended even in symptomatic HIV-infected adults
and children.47-49 Viral loads may increase after vaccination, but CD4 counts
are unaffected and the benefits exceed the risk.50-53
Hepatitis B vaccine is safe to use, but the immunological response may be
poor. HIV-positive adults should receive 3 doses of the H-B-VAX II dialysis
formulation and HIV-positive children should receive 3 doses of Engerix-B
adult formulation. Antibody level should be measured at the completion
of the vaccination schedule. Because many HIV-positive men who have sex
with men may already have been exposed to the hepatitis B and hepatitis
A viruses, their susceptibility should be determined in order to avoid
unnecessary vaccination.
Susceptible HIV-infected individuals should be vaccinated against hepatitis A.54
BCG must not be given to HIV-infected children or adults because of the risk
of disseminated BCG infection.
Yellow fever and oral live attenuated typhoid vaccines should not be given to
HIV-infected individuals. Vi polysaccharide typhoid, Japanese encephalitis
and rabies vaccines are safe and can be used for the usual indications (see
Chapter 2.2, Vaccination for international travel).

2.3.3.5 Individuals with functional or anatomical asplenia55,56


Individuals with an absent or dysfunctional spleen are at an increased risk of
fulminant bacteraemia, most notably pneumococcal, for the rest of their lives.57
Pneumococcal vaccination
All individuals with functional or anatomical asplenia should be vaccinated
against invasive pneumococcal disease. In elective splenectomy, the vaccination
should be completed, if possible, 2weeks before the operation; in unplanned
splenectomy, the vaccination should commence when the patient has recovered
from the surgery.58
Children 5years of age with functional or anatomical asplenia should be given
the age-appropriate course of pneumococcal vaccines for medical-risk children
(see Table 3.15.1 Summary table pneumococcal vaccination schedule for children
9years of age and Section 1.3.5, Catch-up).
Children who develop functional or anatomical asplenia between 6 and 9years
of age should be given 2 doses of 7vPCV 2months apart, followed by a dose of
23vPPV 2months later.

100 The Australian Immunisation Handbook 9th Edition

Individuals 10years of age with functional or anatomical asplenia should be


given:
an initial dose of 23vPPV,
revaccination with 23vPPV 5years after the initial dose (of 23vPPV), and
1 further revaccination (third dose) should be given at either 5years after
the first revaccination or at 50years (Indigenous adults) or 65years (nonIndigenous adults) of age, whichever is later.
It should be noted that the above regimens cannot provide prolonged protection
against all invasive pneumococcal disease. It is particularly important that
individuals with functional or anatomical asplenia are informed of the altered
immune status associated with asplenia and the increased life-long risk of
severe bacterial infection, that they should seek urgent medical assessment for
any febrile illness, and that they should always wear a medical alert bracelet or
necklace.
NB. Children 5years of age with splenic dysfunction, most frequently due
to sickle cell disease, should also be treated with daily doses of penicillin V,
commencing before the age of 4months and continuing until 5years of age
(penicillin V 125mg twice daily, increasing to 250mg twice daily when they
reach 4years of age).
Meningococcal vaccination
All individuals 1 year of age with functional or anatomical asplenia should be
vaccinated with a single dose of MenCCV, although the vaccine can be given
from 6weeks of age (see also Chapter 3.12, Meningococcal disease). This should
be followed by a dose of the 4vMenPV at 2years of age (see also Chapter 3.12,
Meningococcal disease). If MenCCV is given, a period of at least 2weeks should
elapse before 4vMenPV is administered. A single revaccination with 4vMenPV is
recommended 3 to 5years later.
Hib vaccination
Children should be up-to-date with Hib vaccination. A single dose of Hib vaccine
is recommended for splenectomised adults.
Adults with conditions such as systemic lupus erythematosus (SLE), rheumatoid
arthritis (RA) and multiple sclerosis (MS) should be given influenza and
pneumococcal polysaccharide vaccines, due to potential morbidity and mortality
from infection, despite the potential for reduced immunogenicity in some
patients (described below).59-61
For conditions such as SLE and RA, theoretical concerns that vaccines may
exacerbate or cause these diseases have not been substantiated, despite a number
of sporadic case reports. There is potential for reduced immunogenicity of
vaccines, due to both immunosuppressive therapies and the underlying disease.62

Groups with special vaccination requirements 101

2.3 Groups with special


vaccination requirements

2.3.3.6 Individuals with autoimmune diseases

Small controlled studies suggest that approximately one-third of patients with


SLE or RA receiving immunosuppressive therapies may mount a lower antibody
response to influenza and pneumococcal vaccines compared with healthy
controls.60,62 A small proportion of patients may mount very little or no response
to the pneumococcal vaccine.60 Importantly, clinical and laboratory measures
of disease activity, and the choice, duration and dose of immunosuppressive
therapies, do not predict who these poor responders will be.60,62,63 In a study
involving 149 patients with RA taking immunosuppressive agents, including
tumour necrosis factor (TNF) blockers, and 47 healthy controls, patients on TNF
blockers showed similar responses to pneumococcal vaccination to controls.63
Patients treated with methotrexate (as monotherapy or with TNF blockers) had a
reduced antibody response to vaccination.63
There is clear evidence that multiple sclerosis is not exacerbated by influenza
vaccination, and either insufficient or no evidence that other vaccines increase
this risk.64

2.3.4 Vaccination of recent recipients of


normal human immunoglobulin
The immune response to live viral vaccines (with the exception of yellow fever
vaccine) may be inhibited by normal human immunoglobulin. The interval
recommended is dependent on the type of immunoglobulin given (see Table 2.3.5
Recommended intervals between either immunoglobulins or blood products and MMR,
MMRV or varicella vaccination).
When hyperimmune globulin is used against a specific infection, such as varicella,
vaccination against other live viruses need not be deferred. Specialist advice
should be sought if high-dose or intravenous immunoglobulins have been used.

2.3.5 Vaccination of patients following receipt of


other blood products including blood transfusions
People who have received a blood transfusion, including mass blood
transfusions, do not require revaccination. However, following the receipt of any
blood product, including plasma or platelets, an interval of 3 to 7months should
elapse, dependent on the blood product transfused, before vaccination with an
MMR, MMRV or varicella vaccine (see Table 2.3.5 Recommended intervals between
either immunoglobulins or blood products and MMR, MMRV or varicella vaccination).
An interval is suggested because there may be low levels of antibodies present in
the blood product that may impair the immune response to the live vaccine.

102 The Australian Immunisation Handbook 9th Edition

Table 2.3.5: Recommended intervals between either immunoglobulins or blood


products and MMR, MMRV or varicella vaccination65
Route

Blood transfusion:

Washed RBCs

Dose
IU ormL

Estimated
mg IgG/kg

Interval
(months)

IV

10mL/kg

Negligible

RBCs, adenine-saline
added

IV

10mL/kg

10

Packed RBCs

IV

10mL/kg

2060

Whole blood

IV

10mL/kg

80100

Cytomegalovirus immunoglobulin

IV

3mL/kg

150

Hepatitis A prophylaxis
(as NHIG)

IM

0.5mL (<25kg)

1.0mL (2550kg)
2.0mL (>50kg)

Hepatitis B prophylaxis
(as HBIG)

IM

ITP (as NHIG [Intravenous])


ITP (as NHIG [Intravenous])
ITP or Kawasaki disease
(as NHIG [Intravenous])

100 IU

10

IV

400

IV

1000

10

IV

16002000

11

400 IU

(max. dose 15mL)

Measles prophylaxis (as NHIG):


Standard

IM

0.2mL/kg

Immunocompromised

IM

0.5mL/kg

Plasma or platelet products

IV

10mL/kg

160

Rabies prophylaxis (as RIG)

IM

20 IU/kg

22

Replacement (or therapy) of immune


deficiencies (as NHIG [Intravenous],
various doses)

IV

300400

Rh (D) IG (anti-D)

IM

Tetanus (as TIG for IM use)

IM

IM

0
250 IU (given within
24 hrs of injury)

10

500 IU (>24 hrs after


injury)

20

200 IU (010kg)

400 IU (1130kg)
600 IU (>30kg)

Groups with special vaccination requirements 103

2.3 Groups with special


vaccination requirements

Varicella prophylaxis (as ZIG)

2.3.6 Vaccination of patients with bleeding disorders


Intramuscular injection may lead to haematoma formation in patients with
disorders of haemostasis, and to pressure necrosis, muscle contractures or
nerve compression in patients with severe coagulopathies. Children with
inherited coagulopathies should receive factor replacement before intramuscular
injection. Unless warfarin doses are known to be stable, patients receiving this
anticoagulant should have prothrombin times measured before intramuscular
injections, which should be deferred if the INR (international normalised ratio) is
greater than 3.0. Patients with platelet counts of less than 50 x 109/L should not
receive intramuscular injections.
The subcutaneous route could be considered as an alternative to the
intramuscular route in patients with bleeding disorders; seek expert advice.19

2.3.7 Vaccination before or after anaesthesia/surgery


Recent or imminent surgery is not a contraindication to vaccinations and recent
vaccination is not a contraindication to surgery (see Section 1.3.4, Pre-vaccination
screening). There are no randomised controlled trials providing evidence of
adverse outcomes with anaesthesia and surgery in recently vaccinated children.
It is possible that the systemic effects from recent vaccination, such as fever and
malaise, may cause confusion in the post-operative period. Elective surgery
and anaesthesia may be postponed for 1week after inactive vaccination and
for 3weeks after live attenuated viral vaccination in children, and routine
vaccination may be deferred for 1week after surgery.66
A patient who receives any blood products during surgery will need to be
informed of the need to delay any vaccinations (see Table 2.3.5 Recommended
intervals between either immunoglobulins or blood products and MMR, MMRV or
varicella vaccination).

2.3.8 Vaccination of those at occupational risk


Certain occupations, particularly those associated with healthcare, are associated
with an increased risk of some vaccine-preventable diseases.67,68 Furthermore,
some infected workers, particularly healthcare workers and childcare workers,
may transmit infections such as influenza, rubella, measles, mumps, varicella and
pertussis to susceptible contacts with the potential for serious health outcomes.
Many infectious diseases, measles in particular, are highly infectious severaldays
before symptoms become apparent.
Where workers are at significant occupational risk of acquiring a vaccinepreventable disease, the employer should implement a comprehensive occupational
vaccination program which includes a vaccination policy, current staff vaccination
records, provision of information about the relevant vaccine-preventable diseases,
and the management of vaccine refusal (which should, for example, include
reducing the risk of a healthcare worker (HCW) transmitting disease to a vulnerable

104 The Australian Immunisation Handbook 9th Edition

patient). Employers should take all reasonable steps to encourage non-immune


workers to be vaccinated.
Current recommended vaccinations for people at risk of occupationally acquired
vaccine-preventable diseases are listed in Table 2.3.6.
Standard precautions should be adopted where there is risk of occupational
exposure to blood and body fluids. Preventive measures include the appropriate
handling and disposal of sharps, and the donning of gloves, when handling body
fluids, and goggles/face shields, when splashes are likely.
If a non-immune person is exposed to a vaccine-preventable disease, postexposure prophylaxis should be administered where indicated.
Table 2.3.6: Recommended vaccinations for those at risk of occupationally
acquired vaccine-preventable diseases*
OCCUPATION

DISEASE/VACCINE

HEALTHCARE WORKERS (HCW)


All HCW:

Hepatitis B

including all workers and students directly involved


in patient care or the handling of human tissues

Influenza
Pertussis (dTpa,
provided dTpa has not
been given previously)
MMR (if non-immune)
Varicella (if seronegative)

HCW who work with remote Indigenous communities


in NT, QLD, SA and WA; medical, dental and nursing
undergraduate students (in some jurisdictions)

Vaccines listed for All


HCW, plus hepatitis A

HCW who may be at high risk of exposure to


drug-resistant cases of tuberculosis

Vaccines listed for All


HCW, plus BCG

THOSE WHO WORK WITH CHILDREN


All those working with children including:
Childcare and preschool staff (including childcare students)

School teachers (including student teachers)

MMR (if non-immune)


Varicella (if seronegative)

Outside school hours carers


Child counselling services workers
Youth services workers
Childcare and preschool staff

Vaccines listed for


All those working
with children plus
hepatitis A vaccine

Groups with special vaccination requirements 105

2.3 Groups with special


vaccination requirements

Correctional staff working where infants/


children cohabitate with mothers

Pertussis (dTpa,
provided dTpa has not
been given previously)

OCCUPATION

DISEASE/VACCINE

CARERS
Carers of people with intellectual disabilities

Hepatitis A
Hepatitis B

Staff of nursing homes and long-term care facilities

Influenza

Providers of home care to people at risk of high influenza morbidity Influenza


EMERGENCY AND ESSENTIAL SERVICE WORKERS
Police and Emergency Workers

Hepatitis B, influenza

Armed Forces personnel

Hepatitis B, influenza
(and other vaccines
relevant to deployment)

Staff of correctional facilities

Hepatitis B, influenza

LABORATORY PERSONNEL
Laboratory personnel handling veterinary specimens or
working with Q fever organism (Coxiella burnetii)

Q fever

Laboratory personnel handling either bat


tissues or ABL or rabies virus

Australian bat lyssavirus


(ABL) and rabies

Laboratory personnel routinely working


with other infectious agents

Anthrax
Vaccinia poxviruses
Poliomyelitis
Typhoid
Yellow fever
Meningococcal disease
Japanese encephalitis

WORKING WITH SPECIFIC COMMUNITIES


Workers who live with or make frequent visits to remote
Indigenous communities in NT, QLD, SA and WA

Hepatitis A

Workers assigned to the outer Torres Strait Islands


for amonth or more during the wet season

Japanese encephalitis

106 The Australian Immunisation Handbook 9th Edition

OCCUPATION

DISEASE/VACCINE

WORKING WITH ANIMALS


Veterinarians, veterinary students, veterinary nurses

Q fever
Australian bat lyssavirus
(ABL) and rabies

Agricultural college staff and students exposed to high-risk animals Q fever


Abattoir workers and contract workers in
abattoirs (excluding pig abattoirs)

Q fever

Livestock transporters
Sheep shearers and cattle, sheep and dairy farmers
Those culling/processing kangaroos or camels
Tanning and hide workers
Goat farmers
Livestock saleyard workers
Those handling animal products of conception
Those who come into regular contact with bats
(both flying foxes and microbats), bat-handlers,
bat scientists, wildlife officers, zoo curators

Australian bat lyssavirus


(ABL) and rabies

Poultry workers, and others handling poultry, including those who


may be involved in culling during an outbreak of avian influenza

Influenza

OTHERS EXPOSED TO HUMAN TISSUE, BLOOD, BODY FLUIDS OR SEWAGE


Embalmers

Hepatitis B, BCG

Sex industry workers

Hepatitis A
Hepatitis B

Workers who perform skin penetration


procedures, eg. tattooists, body-piercers

Hepatitis B

Funeral workers and other workers who have regular contact with
human tissue, blood or body fluids and/or used needles or syringes

Hepatitis B

Plumbers or other workers in regular contact with untreated sewage Hepatitis A

All adults born during or since 1966 should have evidence of either receiving 2 doses
of MMR vaccine or immunity. Adults born before 1966 are considered to be immune due
to extensive measles circulating widely in the community during this period of time (see
Chapter 3.11, Measles).
People with a repeated risk of exposure or working with large quantities or concentrations
of Bacillus anthracis cultures. For information regarding anthrax vaccination, please contact
the Office of Health Protection in the Australian Government Department of Health and
Ageing, Canberra.

Groups with special vaccination requirements 107

2.3 Groups with special


vaccination requirements

* Work activities, rather than job title, should be considered on an individual basis to ensure
an appropriate level of protection is afforded to each worker.

2.3.9 Vaccination of immigrants to Australia69


Vaccination status is not routinely assessed in children and adults entering
Australia as refugees or immigrants. They may be incompletely vaccinated
according to the Australian schedule or have incomplete records of vaccination.
The World Health Organization website www.who.int/countries/en lists
immunisation schedules for most countries and may provide some information
regarding vaccine schedules.
If an immigrant has no valid documentation of vaccination, the standard
catch-up schedule should be commenced. Serological testing to determine
the need for specific vaccinations is not recommended in the absence of
documented vaccination. If a child is 12months of age, the first doses of
DTPa, hepatitis B, IPV, MMR, MenCCV, 7vPCV and Hib vaccines can be
given at the same visit. For details, see Section 1.3.5, Catch-up.
If there is a valid record of vaccination, the history of previous doses should
be taken into account when planning a catch-up vaccination schedule.
Immigrant adults need to be targeted for vaccination, especially against
rubella using MMR. This is particularly important for women of childbearing age.
All vaccines administered to children <7years of age should be documented
on the ACIR, including those for children not enrolled with Medicare and
vaccinations documented pre-arrival.
ACIR History Statements can be issued after documentation of overseas
vaccination(s) have been recorded on ACIR.
The Australian Government Department of Immigration and Citizenship
(DIAC) may in some circumstances be able to provide information regarding
vaccine(s) administered to refugees before entering Australia.

2.3.10 Vaccination of inmates of correctional facilities


Inmates of correctional facilities are at risk of acquiring influenza, hepatitis A and
hepatitis B, and should be vaccinated against these infections (see Chapter 3.5,
Hepatitis A, Chapter 3.6, HepatitisB and Chapter 3.9, Influenza).70,71

2.3.11 Vaccination of men who have sex with men


Men who have sex with men are at risk of acquiring hepatitis A and hepatitis B,
and should be vaccinated against these infections (see Chapter 3.5, Hepatitis A
and Chapter 3.6, Hepatitis B).

108 The Australian Immunisation Handbook 9th Edition

2.3.12 Vaccination of injecting drug users


Injecting drug users are at risk of acquiring hepatitis A and hepatitis B, and
should be vaccinated against these infections (see Chapter 3.5, Hepatitis A and
Chapter 3.6, Hepatitis B).

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

2.3 Groups with special


vaccination requirements

Groups with special vaccination requirements 109

PART 3: Vaccines listed by disease


3.1 Australian bat lyssavirus
infection and rabies
Virology
Australian bat lyssavirus (ABL) and rabies virus are members of the family
Rhabdoviridae, genus Lyssavirus. There are 7 known genotypes within the genus
Lyssavirus; ABL (genotype 7) is more closely related to rabies virus (genotype 1)
than any of the other 6 genotypes.

Clinical features
Based on the 2 recognised human cases of ABL infection, it has to be assumed
that ABL has the same clinical features as rabies. The incubation period of
rabies is usually 3 to 8weeks, but can range from as short as aweek to, on rare
occasions, severalyears. The risk of rabies is higher, and the incubation period
shorter, after severe and multiple wounds proximate to the central nervous
system (such as on the head and neck) and in richly innervated sites (such as the
fingers).
Typically, in the prodromal phase of rabies, which lasts up to 10days, the patient
may experience non-specific symptoms such as anorexia, cough, fever, headache,
myalgia, nausea, sore throat, tiredness and vomiting.1 Paraesthesiae and/
or fasciculations at or near the site of the wound may be present at this stage.
Anxiety, agitation and apprehension may also occur.
Most rabies patients present with the furious or encephalitic form.1 In the
encephalitic phase, objective signs of nervous system involvement include
aerophobia, hydrophobia, bizarre behaviour, disorientation and hyperactivity.
Signs of autonomic instability such as hypersalivation, hyperthermia and
hyperventilation may occur.1 The neurological status of the patient deteriorates
over a period of up to 12days, and the patient either dies abruptly from cardiac
or respiratory arrest, or lapses into a coma. Rabies is almost invariably fatal.

Epidemiology
Rabies is endemic throughout much of Africa, Asia, the Americas and Europe,
where the virus is maintained in certain species of mammals.1 Australia, New
Zealand, Japan, Papua New Guinea and Pacific Island nations are free of
endemic rabies. Human rabies characteristically follows a bite from a rabid
animal, most frequently a dog, but in some parts of the world, other animals,
such as jackals and bats, are important sources of exposure. In countries where
rabies vaccination of domestic animals is widespread (North America and
Europe), wild animals such as raccoons and foxes are important reservoirs.1

110 The Australian Immunisation Handbook 9th Edition

Although rabies in travellers is rare, such cases always fatal continue to be


reported in the medical literature.4,5 Travellers to rabies-endemic regions should
be advised of the risk and to avoid close contact with either wild or domestic
animals; this is particularly important for children. They should be advised
about pre-travel (ie. pre-exposure) rabies vaccination (or, if appropriate, booster
doses), and they should be advised on what to do should they be either bitten or
scratched by an animal while abroad.
In Australia, 2 cases of a fatal rabies-like illness caused by ABL have been
reported, one in 1996 and the other in 1998.6 Both patients had been bitten
by bats. Evidence of ABL infection has since been identified in all 4 species
of Australian fruit bats (flying foxes) and in several species of Australian
insectivorous bats. It should therefore be assumed that all Australian bats have
the potential to be infected with ABL.

Rabies vaccine
Mrieux Inactivated Rabies Vaccine Sanofi Pasteur Pty Ltd. Each 1.0mL
monodose vial of lyophilised vaccine contains at least 2.5 IU inactivated
rabies virus; 100150g neomycin; 70mg human serum albumin; trace of
phenol red (indicator). 1.0mL distilled water as diluent.
Rabipur Inactivated Rabies Virus Vaccine CSL Biotherapies/Novartis
Vaccines. Each 1.0mL monodose vial of lyophilised vaccine contains at least
2.5 IU inactivated rabies virus; trace amounts of neomycin, chlortetracycline
and amphotericin B; may contain trace amounts of bovine gelatin. May
contain traces of egg protein. 1.0mL distilled water as diluent.
The Mrieux vaccine is a lyophilised, stabilised suspension of inactivated Wistar
rabies virus that has been cultured on human diploid cells and then inactivated
by beta-propiolactone. This human diploid cell vaccine (HDCV) is coloured offwhite, but after reconstitution with the diluent it turns a pinkish colour due to
the presence of phenol red. The vaccine does not contain a preservative.
Rabipur is a lyophilised, stabilised suspension of inactivated Flurey LEP rabies
virus that has been cultured on purified chick embryo cells and then inactivated
by beta-propiolactone. This purified chick embryo cell vaccine (PCECV) does not
contain a preservative.
The above two vaccines, and other tissue culture vaccines, are interchangeable.

Australian bat lyssavirus infection and rabies 111

3.1 Australian bat


lyssavirus infection
and rabies

Cases of rabies after animal scratches, the licking of open wounds or saliva
contact with intact mucous membranes are very rare.2,3 Cases have been recorded
after exposure to aerosols in a laboratory and in caves infested with rabid bats,
and cases have been reported following tissue transplantation from donors who
died with undiagnosed rabies.1

Rabies immunoglobulin
Imogam Rabies Sanofi Pasteur Pty Ltd (human rabies immunoglobulin).
Each 1.0mL contains IgG class human rabies antibodies with a minimum
titre of 150 IU; 22.5mg glycine; 1mg sodium chloride. It is supplied in 2mL
and 10mL vials.
Human rabies immunoglobulin (HRIG) is prepared by cold ethanol fractionation
from the plasma of hyperimmunised human donors.

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.7 Rabies
vaccine, diluent and HRIG should be transported and stored at +2C to
+8C. Do not freeze. Reconstituted vaccine should be used immediately after
reconstituting. The HRIG should be used immediately once the vial is opened.

Dosage and administration


NB. The doses of rabies vaccines are the same for both children and adults.

(i) Pre-exposure prophylaxis


The dose of rabies vaccine for pre-exposure prophylaxis is 1.0mL by IM injection,
ondays 0, 7 and 28. (HDCV can also be given by the subcutaneous (SC) route.)

(ii) Post-exposure treatment


The dose of rabies vaccine for post-exposure treatment is 1.0mL by IM injection,
ondays 0, 3, 7, 14 and 2830. (HDCV can also be given by the SC route.) The dose
of HRIG is 20 IU/kg body mass by infiltration around the wounds; the remainder
of the dose should be administered by IM injection.

Recommendations
(i) Pre-exposure prophylaxis for Australian bat lyssavirus infection and rabies
Rabies vaccine is effective and safe when used for pre-exposure prophylaxis
for rabies.8 Although data on the effectiveness of rabies vaccine as prophylaxis
against ABL infection are limited, the available animal data9 and clinical
experience support its use. Pre-exposure prophylaxis simplifies the management
of a subsequent exposure because fewer doses of vaccine are needed and
because HRIG is not required. (Rabies immunoglobulin is often difficult, or even
impossible, to obtain in many developing countries.)

112 The Australian Immunisation Handbook 9th Edition

Pre-exposure prophylaxis with rabies vaccine is recommended for:

expatriates and travellers who will be spending prolonged periods (ie. more
than amonth) in rabies-endemic areas. (NB. This time interval, of more than
amonth, is arbitrary, and rabies has occurred in travellers following shorter
periods of travel),5
people working with mammals in rabies-endemic areas, and
research laboratory personnel working with live lyssaviruses.
Pre-exposure prophylaxis for both ABL infection and rabies, for all ages, consists
of a total of 3 IM (IM or SC if HDCV is used) injections of 1mL of rabies vaccine,
the second given 7days after the first, and the third given 28days after the first.
Although the third dose can be given at 21days,1 there are no data to support
the use of an even more accelerated schedule for those with limited time before
travel to a rabies endemic area.
Doses should be given in the deltoid area, as rabies neutralising antibody titres
may be reduced after administration in other sites. In particular, vaccine should
never be given in the buttock, as failure of pre-exposure prophylaxis has been
reported when given by this route.
Because the antibody response is reported as satisfactory after the pre-exposure
prophylaxis regimen, routine serological testing to confirm seroconversion is not
necessary. However, people with impaired immunity who are at risk of exposure
to ABL or rabies should have their antibody titres determined 2 to 3weeks after
the third dose of vaccine.
Booster doses of rabies vaccine are recommended for immunised people who
have ongoing exposure to either ABL or rabies. People who work with live
lyssaviruses in research laboratories should have rabies antibody titres measured
every 6months. If the titre is reported as inadequate (<0.5 IU/mL), they should
have a booster dose. Others with occupational exposures to bats in Australia,
and those who are likely to be exposed to potentially rabid animals in endemic
countries, should have rabies antibody titres measured every 2years. If the titre is
reported as inadequate, they should have a booster dose. Alternatively, a booster
dose may be offered every 2years without determining the antibody titre.

Intradermal pre-exposure prophylaxis


There are no data on the protection provided by intradermal (ID) rabies
vaccination for the prevention of ABL infection. Therefore, ID pre-exposure
administration of rabies vaccine should not be used for pre-exposure prophylaxis of ABL.
Antibody titres are lower and wane more rapidly after ID compared to either IM
or SC administration of rabies vaccine, and there may be a slow initial immune

Australian bat lyssavirus infection and rabies 113

3.1 Australian bat


lyssavirus infection
and rabies

people in Australia liable to receive bites or scratches from bats (this includes
bat handlers, veterinarians, wildlife officers and others who come into direct
contact with bats),

response following exposure to rabies virus in those given ID rabies vaccine.10


For these 2 reasons, it is strongly recommended that the IM (IM or SC if HDCV is
used) route be used for pre-exposure prophylaxis.
However, the cost of IM (IM or SC if HDCV is used) rabies vaccination may be
prohibitive for some travellers. In this circumstance, ID rabies vaccination, using
a dose of 0.1mL ondays 0, 7 and 28, may be considered, provided that:
it is given by those with not only expertise in, but also regular practice of, the
ID technique,
it must not be administered to anyone known to have impaired immunity,
it must not be administered to those taking either chloroquine or other
antimalarials structurally related to chloroquine (eg. mefloquine) at either the
time of, or within amonth following, vaccination,
any remaining vaccine is discarded at the end of the session during which the
vial is opened, and
the rabies antibody level should be checked 2 to 3weeks following
completion of the pre-exposure course of ID vaccine.
The use of the ID route for rabies vaccination is the practitioners own
responsibility, as rabies vaccines are not licensed for use via this route in Australia.
The ID route should never be used to administer rabies vaccine by practitioners who
only occasionally provide travel medicine services.

(ii) Post-exposure treatment for Australian bat lyssavirus and rabies exposures
Rabies vaccine and HRIG are effective and safe when used for post-exposure
treatment following rabies exposures. Although data on the effectiveness of
rabies vaccine and HRIG as post-exposure treatment against ABL infection
are limited, the available animal data9 and clinical experience support its use.
The essential components of post-exposure treatment for either ABL or rabies
exposures are prompt local wound management and, for people who have not
previously been vaccinated, administration of HRIG and rabies vaccine as soon
as is practicable.8 Both HRIG and rabies vaccine are available for post-exposure
treatment from the relevant State/Territory health authorities (see Appendix 1,
Contact details for Australian, State and Territory Government health authorities and
communicable disease control).
Post-exposure treatment should be considered whenever a bite, scratch or
mucous membrane exposure to saliva from any Australian bat has occurred,
regardless of the extent of the bite or scratch, the time lapsed since the exposure,
the species of bat involved, and even if the bat was apparently normal in
appearance and behaviour. (Although ABL is more likely to be found in bats that
either appear unwell or are behaving abnormally,11 it has to be assumed that any
bat is potentially infected with ABL.)

114 The Australian Immunisation Handbook 9th Edition

However, exposure to bat blood, urine or faeces, or to a bat that has been dead
for more than 4 hours, does not warrant post-exposure treatment.

The relevant State/Territory health authority should be contacted about any


animal bite or scratch sustained in a rabies-endemic area. Dogs and monkeys
comprise the usual exposures in Asia, Africa and Central and South America,
but exposures to other mammals must also be assessed for potential rabies
transmission. If a traveller presents >10days after being bitten or scratched by
either a dog or cat in an endemic country, and it can be reliably ascertained that
the animal has remained healthy (>10days after the exposure), post-exposure
treatment is not required;8,12 otherwise, a complete course of treatment should
be administered, even if there has been a considerable delay in reporting the
incident.
Immediate and thorough washing of all bite wounds and scratches with soap
and water, and the application of a virucidal preparation such as povidoneiodine solution after the washing, is an important measure in the prevention of
ABL infection and rabies.1 Consideration should be given at this stage of wound
management to the possibility of tetanus and other wound infections, and
appropriate measures taken. Primary suture of a bite from a potentially rabid
animal should be avoided. Bites should be cleaned, debrided and well infiltrated
with HRIG (see below).

a) Use of rabies vaccine in post-exposure treatment


Following the local wound management, the subsequent post-exposure
treatment for either ABL or rabies exposures consists of: (i) a total of 5 doses of
1.0mL of rabies vaccine given by IM (IM or SC if HDCV is used) injection; and
(ii) HRIG (see below).
The volume of rabies vaccine administered to infants and children is the same
as that given to adults (ie. 1.0mL). The first dose of vaccine is given as soon
as is practicable (day 0), and subsequent doses are given ondays 3, 7, 14 and
2830; deviations of a fewdays from this schedule are probably unimportant.8 In
adults and children, the vaccine should be administered into the deltoid area, as
administration in other sites may result in reduced neutralising antibody titres.

Australian bat lyssavirus infection and rabies 115

3.1 Australian bat


lyssavirus infection
and rabies

Where post-exposure treatment for a potential exposure to ABL is indicated,


the bat should, if possible, without placing others at risk of exposure, be
kept and arrangements promptly made for testing by the relevant State/
Territory veterinary or health authority. Following the wound management,
the administration of HRIG and rabies vaccine can be withheld if the result
(concerning the bats ABL status) will be available within 48 hours of the
exposure; if the result will not be available within 48 hours, full post-exposure
treatment should begin as soon as is practicable. Where a bat is tested at a
reference laboratory and later found to be negative for ABL, then post-exposure
treatment for individuals exposed to that bat can be discontinued.

In infants <12months of age, administration into the anterolateral aspect of the


thigh is recommended.
Serological testing to measure response is unnecessary except in unusual
circumstances, such as when the patient is known to have impaired immunity.
In such cases, the antibody titre should be measured 2 to 3weeks after the dose
given at 2830days and a further dose given if the titre is reported as inadequate.

b) Use of rabies immunoglobulin in post-exposure treatment


Rabies has occurred in people who have received post-exposure rabies vaccine
without rabies immunoglobulin being infiltrated in and around the wound.13
Therefore, post-exposure treatment should always include the infiltration of HRIG
in and around wounds at the same time as the first dose of rabies vaccine, the only
exceptions being people with documented evidence of either completion of the
pre-exposure prophylaxis regimen or adequate rabies antibody titres. These
people should receive vaccine only (see below).
A single dose of HRIG is given to provide localised anti-rabies antibody
protection while the patient responds to the rabies vaccine. It should be given
at the same time as the first post-exposure dose of vaccine (day 0). If not given
with the first vaccine dose, it may be given up to day 7, but should not be given
any later in the vaccination course. From day 8 onwards, an antibody response to
rabies vaccine is presumed to have occurred.
The dose of HRIG for all age groups is 20 IU perkg body mass. HRIG should be
infiltrated in and around all wounds using as much of the calculated dose as possible,
and the remainder administered intramuscularly at a site away from the injection
site of rabies vaccine. If the wounds are severe and the calculated volume of
HRIG is inadequate for complete infiltration of all wounds (eg. extensive dog
bites in a young child), the HRIG should be diluted in saline to make up an
adequate volume for the careful infiltration of all wounds.
However, many bat bites occur as small puncture wounds on the fingers;8 such
exposures are probably high-risk exposures because of the extensive nerve
supply to the fingers and hand. Therefore, although infiltration of HRIG into
finger wounds is likely not only to be technically difficult but also to be painful
for the recipient, it must be undertaken. As much of the calculated dose of HRIG
as possible should be infiltrated into finger and hand wounds using either a
25 or 26 gauge needle. To avoid the development of a compartment syndrome,
the HRIG should be infiltrated very gently, and should not cause the adjacent
finger tissue to go frankly pale or white. If necessary, a ring-block using a local
anaesthetic may be required.

116 The Australian Immunisation Handbook 9th Edition

Table 3.1.1: Summary of Australian bat lyssavirus and rabies


post-exposure treatment for non-immune individuals
Immediate (Day 0)

Local treatment

Thorough wound cleansing

Follow-up

Rabies vaccine

1.0mL

1.0mL ondays 3, 7, 14, 2830

Human rabies
immunoglobulin
(150 IU/mL)

20 IU/kg no later than


7days after the first
rabies vaccine dose

Do not give later than


7days after the first
rabies vaccine dose

c) Post-exposure treatment of previously vaccinated people


People who have either completed a recommended course of pre-exposure
prophylaxis, or previous post-exposure treatment, or who have documented
adequate rabies neutralising antibodies, require a modified post-exposure
treatment regimen if potentially exposed to either rabies virus or ABL. Local
wound management as described above must be carried out, and a total of 2
doses of rabies vaccine (1.0mL each) should be given by IM (IM or SC if HDCV
is used) injection on day 0 and day 3. HRIG is not necessary in these cases.
In cases where the vaccination status is uncertain because the documentation
of a full course of rabies vaccine is not available, the standard post-exposure
treatment regimen (HRIG plus 5 doses of rabies vaccine) should be administered.

d) Post-exposure treatment commenced overseas


Australians travelling abroad who are exposed to a potentially rabid animal
may be given post-exposure treatment with vaccines not available in Australia.
However, it is very likely that they will receive a cell culture derived vaccine,
all of which (including both vaccines available in Australia) are considered
interchangeable.14
Therefore, if a person has received a cell culture-derived vaccine abroad, the
standard post-exposure treatment regimen should be continued in Australia with
either HDCV or PCECV. If the post-exposure treatment was started overseas
but HRIG was not given, and the person presents in Australia within 7days
of commencing post-exposure treatment, HRIG should be given as soon as is
practicable (and within 7days of the first rabies vaccine). If the person presents
in Australia 8days or more after commencing post-exposure treatment, then
HRIG should be withheld.

Contraindications
There are no contraindications to post-exposure treatment in a person with a
possible exposure to either ABL or rabies.
A person with an anaphylactic sensitivity to eggs, or to egg proteins, should not
receive PCECV; HDCV should be used instead.

Australian bat lyssavirus infection and rabies 117

3.1 Australian bat


lyssavirus infection
and rabies

Treatment

Adverse events
Cell culture-derived vaccines are generally well tolerated. In a large study, the
following adverse events were reported after administration of HDCV to adults:
sore arm (15 to 25% very common), headache (5 to 8% common), malaise, nausea
or both (2 to 5% common); and allergic oedema (0.1% uncommon).14 Similar
adverse event profiles have been reported for the PCECV; these reactions occur at
the same rates in children.14
Although anaphylactic reactions are rare (approximately 1 per 10000
vaccinations) following administration of HDCV, approximately 6% (common)
of people receiving booster doses may experience allergic reactions.14 The
reactions typically occur 2 to 21days after a booster dose, and are characterised
by generalised urticaria, sometimes with arthralgia, arthritis, oedema, nausea,
vomiting, fever and malaise. These reactions are not life-threatening; they have
been attributed to the presence of beta-propiolactone-altered human albumin in
the implicated vaccines.14 NB. HDCV contains human albumin, whereas PCECV
does not.

Management of adverse events


Once initiated, rabies prophylaxis should not be interrupted or discontinued
because of local reactions or mild systemic reactions. Such reactions can usually
be managed with simple analgesics.
Because ABL infection and rabies are lethal diseases, the recommended
vaccination regimens, in particular the post-exposure treatment regimen, should
be continued even if a significant allergic reaction occurs following a dose of
rabies vaccine. Antihistamines can be administered in an attempt to ameliorate
any subsequent reactions. A patients risk of developing either ABL infection or
rabies must be carefully considered before deciding to discontinue vaccination.

Use of steroids and immunosuppressive agents


Corticosteroids and immunosuppressive agents can interfere with the
development of active immunity and, therefore, if possible, should not be
administered during post-exposure treatment. A person who either has an
immunosuppressing illness or is taking immunosuppressant medications should
have his/her rabies antibody titres checked 2 to 4weeks after completion of the
vaccination regimen (see above).

Use in pregnancy
Pregnancy is never a contraindication to rabies vaccination. Follow-up of 202
Thai women vaccinated during pregnancy did not indicate either increased
medical complications or birth defects.15

118 The Australian Immunisation Handbook 9th Edition

Variations from product information

The HDCV product information recommends a routine sixth dose at 90days in


the post-exposure treatment regimen. This dose is not considered necessary on
a routine basis but a further dose should be offered to a person with impaired
immunity who has an inadequate antibody level following the standard regimen.
It also recommends a pre-exposure booster after a year; boosters are usually
recommended in Australia after 2years (see above).

Rabies in Indonesia
No cases of Bali-acquired rabies have ever been reported in the medical literature
despite many people being bitten and scratched by animals in Bali every year.
Therefore, post-exposure treatment following animal bites sustained in Bali is
currently not warranted, but obviously this situation could change.
However, rabies still exists in other parts of Indonesia including the islands of
Flores, Sulawesi, Sumatra, Ambon and Kalimantan. Post-exposure treatment is
necessary for any animal bite or scratch sustained in any of these locations. Any
doubts or concerns about the need for post-exposure treatment following animal
bites should be discussed with the State/Territory public health authority.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

Australian bat lyssavirus infection and rabies 119

3.1 Australian bat


lyssavirus infection
and rabies

Neither of the product information sheets (of the 2 vaccines available in


Australia) mentions that they can be used for both pre-exposure prophylaxis and
post-exposure treatment for ABL exposures.

3.2 Cholera
Bacteriology
Vibrio cholerae is a motile, curved Gram-negative bacillus and differences in the
O antigens have led to the description of more than 150 serogroups, only two
of which have been found to cause cholera. Cholera is caused by enterotoxin
producing V. cholerae of serogroups O1 and O139 (sometimes referred to as the
Bengal strain). Serogroup O1 includes 2 biotypes (classical and El Tor), each of
which includes organisms of Inaba, Ogawa and Hikojima serotypes. The ability
of V. cholerae to persist in water is determined by the temperature, pH, salinity
and availability of nutrients; it can survive under unfavourable conditions in a
viable dormant state.1 Transmission predominantly occurs when people ingest
faecally contaminated food or water.

Clinical features
Cholera is an acute bacterial infection that is generally characterised by the
sudden onset of painless, profuse, watery diarrhoea. If untreated, more than half
the severe cases will die. Mild cases also occur, as does subclinical infection.1
The cholera toxin does not produce intestinal inflammation. The cholera toxin
induces secretion of increased amounts of electrolytes into the intestinal lumen,
resulting in mild to severe dehydration and, in some cases, metabolic acidosis.

Epidemiology
Cases of cholera in Australia (about 2 to 6 cases a year) almost always occur in
individuals who have been infected in endemic areas of Asia, Africa, the Middle
East, South America or parts of Oceania.2-4 The disease is usually transmitted
via food and water contaminated with human excreta. Shellfish obtained from
contaminated waters have also been responsible for outbreaks.1
In 1977, a locally acquired case led to the discovery of V. cholerae in some rivers
of the Queensland coast.5 Because of this, health workers should be aware that
sporadic cases of cholera may, on rare occasions, follow contact with estuarine
waters.
As the incubation period of the disease may extend up to 5days, surveillance
of household contacts or those exposed to a possible common source should be
maintained for 5days from the date of last exposure. Stool cultures (cultured
using specific media) may be taken from close contacts if required. Food handlers
should not be allowed to return to work until 2 consecutive stool samples, taken
at least 24 hours apart, are negative. Contacts should also be advised to maintain
high standards of personal hygiene to avoid becoming infected. Cases should be
reported immediately to the public health authorities (for contact details, refer

120 The Australian Immunisation Handbook 9th Edition

to Appendix 1, Contact details for Australian, State and Territory Government health
authorities and communicable disease control).

Vaccines
Dukoral Sanofi Pasteur Pty Ltd (inactivated whole-cell V. cholerae O1
in combination with a recombinant cholera toxin B subunit (rCTB)). Each
3.0mL liquid vaccine dose vial contains heat and formalin inactivated Inaba,
Ogawa, classic and El Tor strains of V.cholerae O1, 2.5 x 1010 vibrios of each,
combined with 1.0mg rCTB. The buffer consists of a sachet of effervescent
granules of anhydrous sodium carbonate, sodium bicarbonate, anhydrous
citric acid, sodium citrate, saccharin sodium and raspberry flavour.

Trials of the inactivated vibrio combined with rCTB vaccine have been done
mainly in Bangladesh and Peru.9,13-16 In Bangladesh, a 2-dose regimen showed
protective efficacy of 44% in children 2 to 6years of age and 76% in adults at the
end of 1 year, and 33% and 60%, respectively, after 2years. The studies in Peru
showed an overall efficacy of 61% in 265-year-olds. A recent study undertaken
during a mass oral cholera vaccination program in Mozambique concluded that 1
or more doses of the inactivated oral cholera vaccine was 78% protective.17
To date, there is no vaccine marketed to protect against infection with V. cholerae
O139. A killed oral whole cell cholera bivalent vaccine (against both serogroups
O1 and O139) is currently being evaluated in Vietnam.18,19
A study in short-term Finnish tourists20 showed that the inactivated oral cholera
vaccine also provided a 60% reduction in diarrhoea caused by heat-labile toxin
producing enterotoxigenic E. coli (LT-ETEC). A study in Bangladesh, an endemic
area, showed 67% protection against LT-ETEC for 3months only.21 It can be
expected that the inactivated vaccine will reduce the proportion of travellers
diarrhoea that is caused by LT-ETEC. Approximately 30 to 40% of travellers to
developing countries contract travellers diarrhoea, with an average of 20% of
cases caused by LT-ETEC; hence, the 60% efficacy of the oral inactivated vaccine
against LT-ETEC could be expected to prevent around 10 to 12% of travellers
diarrhoea.22 However, in Australia this vaccine is only registered for the
prevention of cholera.

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.23 Store in a
refrigerator at +2C to +8C. Do not freeze. Protect from light.

Cholera 121

3.2 Cholera

Trials of the safety, immunogenicity and efficacy of oral vaccines, both killed and
live attenuated, have been carried out in the United States, Bangladesh, Thailand,
Indonesia, Chile, Peru and Switzerland.6-12

Dosage and administration


For adults and children over the age of 6years, Dukoral is administered orally
after dissolving the buffer granules in 150mL of water and adding the vaccine
to the solution. Two doses are required, given a minimum of 1week and up to
6weeks apart. If the second dose is not administered within 6weeks, re-start the
vaccination.
For children aged 26years, Dukoral is administered orally after dissolving
the buffer granules in 150mL of water. Half the solution is then poured away
and the entire content of the vaccine vial is mixed with the remaining 75mL.
Children aged 26years should receive 3 doses of the vaccine. Doses are to
be administered with a minimum interval of 1week between doses up to
a maximum interval of 6weeks. If an interval of more than 6weeks occurs
between any of the doses, re-start the vaccination.
Food and drink should be avoided for 1 hour before and 1 hour after
administration of the inactivated cholera vaccine, as it is acid labile.
The inactivated oral cholera vaccine can be given at the same time as other travel
vaccines. However, there should be an interval of at least 8 hours between the
administration of the inactivated oral cholera and oral typhoid vaccines, as the
buffer in the cholera vaccine may affect the transit of the capsules of oral typhoid
vaccine through the gastrointestinal tract.
Adults and children >6years of age should receive a single booster dose after
2years and children 26years of age should receive a booster dose 6months
after completion of the primary course.

Recommendations
Despite the endemicity of cholera in some countries often visited by Australians,
routine cholera vaccination is not recommended as the risk to travellers is very
low. Careful and sensible selection of food and water is of far greater importance
to the traveller than vaccination.
Immunisation should be considered for people at increased risk of diarrhoeal
disease, such as those with achlorhydria, and for people at increased risk of
severe or complicated diarrhoeal disease, such as those with poorly controlled
or otherwise complicated diabetes, inflammatory bowel disease, HIV/AIDS or
other conditions resulting in impaired immunity, or significant cardiovascular
disease. It could also be considered for humanitarian disaster workers.
Vaccination against cholera is not an official requirement for entry into any
foreign country.

122 The Australian Immunisation Handbook 9th Edition

Contraindications
The only contraindications to the use of cholera vaccine are:
anaphylaxis following a previous dose of the vaccine,
anaphylaxis following any component of the vaccine,
inactivated oral cholera vaccine is not recommended for children
<2years of age.

Precautions
Postpone administration during either an acute febrile illness or acute
gastrointestinal illness with persistent diarrhoea or vomiting, until recovered.

There should be an interval of at least 8 hours between the administration


of the inactivated oral cholera and oral typhoid vaccines, as the buffer in the
cholera vaccine may affect the transit of the capsules of oral typhoid vaccine
through the gastrointestinal tract.

Adverse events
The inactivated oral vaccine is uncommonly (<1%) associated with mild
gastrointestinal disturbances.

Use in pregnancy
There is inadequate information on the use of inactivated oral cholera vaccines
during pregnancy and breastfeeding.24

Variations from product information


None.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

Cholera 123

3.2 Cholera

Although the vaccine is not contraindicated in immune impaired individuals,


including HIV-infected individuals, data on effectiveness in this population is
limited.

3.3 Diphtheria
Bacteriology
Diphtheria is an acute illness caused by toxigenic strains of Corynebacterium
diphtheriae, a Gram- positive, non-sporing, non-capsulate bacillus. The exotoxin
produced by C. diphtheriae acts locally on the mucous membranes of the
respiratory tract or, less commonly, on damaged skin, to produce an adherent
pseudomembrane. Systemically, the toxin acts on cells of the myocardium,
nervous system and adrenals.

Clinical features
The incubation period is 2 to 5days. The disease is communicable for up to 4weeks,
but carriers may shed organisms for longer. Spread is by respiratory droplets
or by direct contact with skin lesions or articles soiled by infected individuals.
Pharyngeal diphtheria is characterised by an inflammatory exudate which forms
a greyish or green membrane in the upper respiratory tract which can cause
acute severe respiratory obstruction. Diphtheria toxin can cause neuropathy and
cardiomyopathy, which may be fatal. The introduction of diphtheria antitoxin in the
1890s reduced the death rate to about 10%, but the mortality has not been further
reduced by the use of antibiotics and other modern treatments.1 Effective protection
against diphtheria is achieved by active immunisation with diphtheria vaccine.

Epidemiology
In the early 1900s, diphtheria caused more deaths in Australia than any other
infectious disease, but increasing use of diphtheria vaccines since World War
II has led to its virtual disappearance.2 The current epidemiology of diphtheria
in Australia is similar to that in other developed countries. Almost all recent
cases in the United Kingdom and the United States have been associated with
imported infections.3 Hence, there is still the possibility of an imported case
occurring in Australia, particularly from developing countries, as occurred in
2001 when a case, acquired in East Timor, was notified in Australia.4 There is now
little possibility of acquiring natural immunity or boosting declining immunity
with subclinical infection. It is therefore important for Australians to retain high
levels of immunity through high vaccination coverage.
Disruption of vaccination programs following the collapse of the Soviet Union
resulted in the re-emergence of diphtheria throughout the Newly Independent
States. From 1991 to 1996, there were more than 140000 cases and more than
4000 deaths.5 Cases also occurred in neighbouring European countries and in
visitors to the area. Mass vaccination eventually brought the epidemic under
control.6,7 This experience illustrates the importance of maintaining high levels of
vaccination coverage against diphtheria.

124 The Australian Immunisation Handbook 9th Edition

Vaccines
Diphtheria toxoid is available in Australia only in combination with tetanus and
other antigens.
The acronym DTPa, using capital letters, signifies child formulations of
diphtheria, tetanus and acellular pertussis-containing vaccines. The acronym
dTpa is used for adolescent/adult formulations which contain substantially
lesser amounts of diphtheria toxoid and pertussis antigens (see formulations).
Formulations for children aged <8years
Infanrix hexa GlaxoSmithKline (DTPa-hepB-IPV-Hib; diphtheriatetanus-acellular pertussis-hepatitis B-inactivated poliomyelitis vaccineHaemophilus influenzae type b (Hib)). The vaccine consists of both a 0.5mL
pre-filled syringe containing 30 IU diphtheria toxoid, 40 IU tetanus toxoid,
25g pertussis toxoid (PT), 25g filamentous haemagglutinin (FHA), 8g
pertactin (PRN), 10g recombinant HBsAg, 40 D-antigen units inactivated
polioviruses type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32
D-antigen units type 3 (Saukett) adsorbed onto aluminium hydroxide/
phosphate; phenoxyethanol as preservative; traces of formaldehyde,
polymyxin and neomycin and a vial containing a lyophilised pellet of 10g
purified Hib capsular polysaccharide (PRP) conjugated to 2040g tetanus
toxoid. The vaccine must be reconstituted by adding the entire contents of the
syringe to the vial and shaking until the pellet is completely dissolved. May
also contain yeast proteins.

Infanrix Penta GlaxoSmithKline (DTPa-hepB-IPV; diphtheria-tetanusacellular pertussis-hepatitis B-inactivated poliomyelitis vaccine). Each
0.5mL pre-filled syringe contains 30 IU diphtheria toxoid, 40 IU tetanus
toxoid, 25g PT, 25g FHA, 8g PRN, 10g recombinant HBsAg, 40
D-antigen units inactivated polioviruses type 1 (Mahoney), 8 D-antigen
units type 2 (MEF-1) and 32 D-antigen units type 3 (Saukett) adsorbed onto
aluminium hydroxide/phosphate; phenoxyethanol as preservative; traces of
formaldehyde, polymyxin and neomycin. May also contain yeast proteins.

Diphtheria 125

3.3Diphtheria

Infanrix-IPV GlaxoSmithKline (DTPa-IPV; diphtheria-tetanus-acellular


pertussis-inactivated poliomyelitis vaccine). Each 0.5mL pre-filled syringe
contains 30 IU diphtheria toxoid, 40 IU tetanus toxoid, 25g PT, 25g FHA,
8g PRN, 40 D-antigen units inactivated polioviruses type 1 (Mahoney),
8 D-antigen units type 2 (MEF-1) and 32 D-antigen units type 3 (Saukett)
adsorbed onto aluminium hydroxide; phenoxyethanol as preservative;
traces of formaldehyde, polymyxin and neomycin.

Formulations for people aged 8years


Adsorbed diphtheria-tetanus vaccine
ADT Booster Statens Serum Institut/CSL Biotherapies (dT; diphtheriatetanus, adult formulation). Each 0.5mL pre-filled syringe or monodose vial
contains 2 IU diphtheria toxoid and 20 IU tetanus toxoid adsorbed onto
0.5mg aluminium hydroxide.
Combination vaccines
Adacel Sanofi Pasteur Pty Ltd (dTpa; diphtheria-tetanus-acellular
pertussis). Each 0.5mL monodose vial contains 2 IU diphtheria toxoid, 20
IU tetanus toxoid, 2.5g PT, 5g FHA, 3g PRN, 5g pertussis fimbriae
(FIM) 2+3; 1.5mg aluminium phosphate; phenoxyethanol as preservative;
traces of formaldehyde.
Adacel Polio Sanofi Pasteur Pty Ltd (dTpa; diphtheria-tetanus-acellular
pertussis-inactivated poliomyelitis vaccine). Each 0.5mL monodose vial
contains 2 IU diphtheria toxoid, 20 IU tetanus toxoid, 2.5g PT, 5g
FHA, 3g PRN, 5g FIM 2+3, 40 D-antigen units inactivated polioviruses
type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32 D-antigen
units type 3 (Saukett); 1.5mg aluminium phosphate; phenoxyethanol
as preservative; traces of formaldehyde, polymyxin, neomycin and
streptomycin.
Boostrix GlaxoSmithKline (dTpa; diphtheria-tetanus-acellular pertussis).
Each 0.5mL monodose vial or pre-filled syringe contains 2 IU diphtheria
toxoid, 20 IU tetanus toxoid, 8g PT, 8g FHA, 2.5g PRN, adsorbed
onto 0.5mg aluminium hydroxide/phosphate; 2.5mg phenoxyethanol as
preservative. May contain traces of formaldehyde.
Boostrix-IPV GlaxoSmithKline (dTpa-IPV; diphtheria-tetanus-acellular
pertussis-inactivated poliomyelitis vaccine). Each 0.5mL pre-filled syringe
contains 2 IU diphtheria toxoid, 20 IU tetanus toxoid, 8g PT, 8g FHA,
2.5g PRN, 40 D-antigen units inactivated polioviruses type 1 (Mahoney),
8 D-antigen units type 2 (MEF-1) and 32 D-antigen units type 3 (Saukett)
adsorbed onto aluminium hydroxide/phosphate; traces of formaldehyde,
polymyxin and neomycin.
Diphtheria vaccination stimulates the production of antitoxin, which protects
against the toxin produced by the organism. The immunogen is prepared by
treating a cell-free preparation of toxin with formaldehyde, thereby converting it
into the innocuous diphtheria toxoid. Diphtheria toxoid is usually adsorbed onto
an adjuvant, either aluminium phosphate or aluminium hydroxide, to increase

126 The Australian Immunisation Handbook 9th Edition

its immunogenicity. Antigens from Bordetella pertussis, in combination vaccines,


also act as an effective adjuvant.
Circulating levels of antitoxin are closely related to protection from diphtheria.
Antitoxin levels of <0.01 IU are poorly protective, 0.01 to 0.1 IU are usually
protective, and titres of >0.1 IU are associated with more certain and prolonged
protection.8 Complete immunisation induces protective levels of antitoxin lasting
throughout childhood but, by middle age, at least 50% of vaccinees have levels
<0.1 IU.9-11 This has been confirmed in Australia by a recent national serosurvey.12
Single low doses of toxoid in previously immunised adults induce protective
levels within 6weeks.13
Production of DT (CDT vaccine), registered for use in children <8years of age,
ceased in June 2005.
ADT Booster can be used for the booster dose of dT in people aged 8years or, if
necessary, for the primary dT course (see Variations from product information).

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.14 Store at
+2C to +8C. Protect from light. Do not freeze.

Dosage and administration


The dose of diphtheria-containing vaccine is 0.5mL by IM injection.
Do not mix DTPa-containing vaccines or dT vaccine with any other vaccine in the
same syringe, unless specifically registered for use in this way.

(i) Vaccination in childhood


The recommended primary course of vaccination is at 2, 4 and 6months of age.
A booster dose of DTPa is given at 4years of age. Immunity to diphtheria will
not be compromised before the booster dose, as the serological response to the
primary course of vaccination is usually sufficient for thoseyears. A second
booster, using the adolescent/adult formulation, dTpa, at 1217years of age, is
essential for maintaining immunity to diphtheria in adults. Vaccination against
diphtheria is part of the National Immunisation Program (NIP) schedule,
diphtheria toxoid being given in combination with tetanus toxoid and acellular
pertussis as DTPa vaccine. Before the 8th birthday, DTPa-containing vaccines
should be given, as they contain a larger dose of diphtheria toxoid. After the 8th
birthday, smaller doses of toxoid (dT or adolescent/adult formulation dTpa)
should be given. Dose reduction is necessary because of the increased incidence
of local and systemic reactions to diphtheria toxoid in older children and adults.
For details on the management of children who have missed doses in the NIP
schedule, see Section 1.3.5, Catch-up.

Diphtheria 127

3.3Diphtheria

Recommendations

(ii) Vaccination of adults


Individuals who have not received any diphtheria vaccines are also likely
to have missed tetanus vaccination. Three doses of dT should be received at
minimum intervals of 4weeks, followed by booster doses at 10 and 20years
after the primary course. It is prudent to give the first of these doses as dTpa, to
also provide boosting to natural immunity from exposure to pertussis, which
is almost universal in unvaccinated adults. In the event that dT vaccine is not
available, dTpa can be used for all primary doses. This is not recommended
routinely because there are no data on the safety, immunogenicity or efficacy of
dTpa in multiple doses for primary vaccination.
All adults who reach the age of 50years without having received a booster dose
of dT in the previous 10years should receive a further booster dose of dT, or
preferably dTpa, if this has not been given previously, to also provide protection
against pertussis.

(iii) Other people at special risk


Diphtheria can be a significant risk for travellers to some countries (particularly
southeast Asia, the Newly Independent States of the former Soviet Union,
Baltic countries or eastern European countries). Travellers to high-risk countries
should receive a booster dose of dT (or dTpa) if they have not received one in the
previous 10years.

Contraindications
The only absolute contraindications to diphtheria vaccine are:
anaphylaxis following a previous dose of the vaccine, or
anaphylaxis following any component of the vaccine.

Adverse events
Mild discomfort or pain at the injection site persisting for up to a fewdays is
common. Uncommon general adverse events following dT vaccine include
headache, lethargy, malaise, myalgia and fever. Acute anaphylactic reactions,
urticaria and peripheral neuropathy very rarely occur (brachial neuritis occurs
in 0.001% of cases). (For specific adverse events following combination vaccines
containing both diphtheria and pertussis antigens, see Chapter 3.14, Pertussis).

The public health management of diphtheria cases


A suspected case of diphtheria is of considerable public health importance, and
should be notified immediately to the State/Territory public health authorities,
who will advise on further management. In general, contacts of a proven or
presumptive diphtheria case will require vaccination (either primary or booster,
depending on vaccination status), and appropriate prophylactic antibiotics.15

128 The Australian Immunisation Handbook 9th Edition

Diphtheria antitoxin and penicillin should be given immediately to suspected


cases. Do not wait for bacteriological confirmation of the disease. Diphtheria
antitoxin derived from horse serum is used because sera of sufficient titre are
not available from humans. Due to the presence of foreign protein, diphtheria
antitoxin may provoke acute, severe, allergic reactions or serum sickness.
Consequently, a test dose should be administered, and if there is evidence of
hypersensitivity, it may be necessary to administer diphtheria antitoxin under
corticosteroid, adrenaline, and antihistamine cover. The therapeutic dose of
antitoxin will depend on the clinical condition of the patient, and may be given
either intramuscularly or diluted for administration in an intravenous infusion.
Expert advice should be sought with respect to antitoxin dose and special
arrangements made if hypersensitivity is suspected. This can be coordinated
through the relevant State/Territory health authority (see Appendix 1, Contact
details for Australian, State and Territory Government health authorities and
communicable disease control).
Diphtheria antitoxin This is currently available only through the Special
Access Scheme.

Use in pregnancy
Refer to Chapter 2.3, Groups with special vaccination requirements, Table 2.3.1
Vaccinations in pregnancy.

Variations from product information

The product information for Infanrix-IPV states that this vaccine may be used as
a booster dose for children 6years of age who have previously been vaccinated
against diphtheria, tetanus, pertussis and poliomyelitis. NHMRC recommends
that booster doses of DTPa and IPV be given at 4years of age; however, this
product may be used for catch-up of the primary schedule or as a booster in
children <8years of age.
The product information for ADT Booster states that this vaccine is indicated
for a booster dose only in children aged 5years and adults who have
previously received at least 3 doses of diphtheria and tetanus vaccines. NHMRC
recommends that, where a dT vaccine is required for any person 8years of
age, ADT Booster can be used, including for primary immunisation against
diphtheria and tetanus.

Diphtheria 129

3.3Diphtheria

The product information for both Infanrix hexa and Infanrix Penta states that
these vaccines may be given as a booster dose at 18months of age. NHMRC
recommends that a booster dose of DTPa (or DTPa-containing vaccines) is not
necessary at 18months of age. However, DTPa-containing vaccine may be used
for catch-up of the primary schedule in children <8years of age.

The product information for adolescent/adult formulations of dTpa-containing


vaccines states that these vaccines are indicated for booster doses only.
NHMRC recommends that, where dT is unavailable for the primary course,
dTpa can be used.
The product information for Adacel and Boostrix (adolescent/adult formulations
of dTpa) states that these vaccines are recommended for use in those aged
>10years. However, NHMRC recommends that they may be used in people
aged 8years. The product information also states that dTpa should not be
given within 5years of a tetanus toxoid-containing vaccine. However, NHMRC
recommends that dTpa vaccines can be administered at any time following
receipt of a diphtheria and tetanus toxoid-containing vaccine.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

130 The Australian Immunisation Handbook 9th Edition

3.4 Haemophilus influenzae type b (hib)


Bacteriology
Haemophilus influenzae is a Gram-negative coccobacillus that is a normal part of
upper respiratory tract flora. Strains isolated from respiratory tract specimens
such as sputum and middle ear or sinus fluid usually do not have a capsule, and
are known as non-typable (NT). Six capsular types (a to f) have been described
and, before the introduction of vaccination against Haemophilus influenzae type
b (Hib), almost all H. influenzae isolates from sterile sites (blood, cerebrospinal
fluid, joint or pleural fluid) were of the b capsular type.
Before Hib immunisation, invasive disease caused by Hib rarely occurred
after the age of 5years. This was because the prevalence of antibody to Hib
progressively increased from the age of 2years, thought to be related to exposure
to Hib (or cross-reacting organisms) colonising the nasopharynx or other sites.
Children <2years of age are usually unable to mount an antibody response to the
type b capsular polysaccharide, even after invasive disease.1

Clinical features
Clinical categories of invasive disease caused by Hib include meningitis,
epiglottitis and a range of other infections such as septic arthritis, cellulitis
and pneumonia. Hib is rarely isolated from the blood without a focal infection
such as the above being evident or developing subsequently. The classical
clinical signs of meningitis neck stiffness and photophobia are often not
detected in infants, who present with drowsiness, poor feeding and high fever.
Epiglottitis (inflammation of the epiglottis) presents with respiratory obstruction,
associated with soft stridor and often drooling in a pale, febrile, anxious child
who remains upright to maximise his or her airway. Meningitis and epiglottitis
are almost invariably fatal without appropriate treatment. There are no specific
clinical features of any of the focal infections due to Hib which enable them
to be differentiated from those due to other organisms. However, before the
introduction of Hib vaccines, epiglottitis was due to Hib in over 95% of cases.2

(i) Before Hib vaccination


Before the introduction of routine Hib vaccination in 1993, there were at least
500 cases of Hib disease in Australian children <6years of age every year, and
a total of 10 to 15 deaths.3 Hib meningitis accounted for approximately 60% of
all invasive Hib disease, most cases occurring in children <18months of age.
The case fatality rate for Hib meningitis was approximately 5%, and up to 40%
of the survivors had neurological sequelae such as deafness and intellectual
impairment.4 Hib epiglottitis was a more common disease presentation than

Haemophilus influenzae type b (Hib) 131

3.4 Haemophilus
influenzae type b (hib)

Epidemiology

in many other countries,5 and usually occurred in children >18months of age.


Other manifestations such as cellulitis, septic arthritis and pneumonia occurred
at a similar age to meningitis.6
The incidence of Hib disease in Aboriginal and Torres Strait Islander children,
especially those in remote and rural areas, was considerably higher than in
non-Indigenous children.7 Most importantly, the onset of Hib disease in this
population was at a much younger age, manifesting mostly as meningitis, with
epiglottitis being rare. Rates of death and long-term morbidity following Hib
meningitis were similar to those observed in non-Indigenous children.7

(ii) After the introduction of Hib vaccination


Since Hib vaccines were included in the routine vaccination schedule in 1993,
there has been a reduction of >90% in notified cases of Hib disease from
502 in 1992 to an average of 30 cases per year between 1999 and 2002, with
approximately 15 cases per year currently reported in Australia (see Figure
3.4.1).8 This reduction has been particularly marked in Indigenous children.9
Similar impressive reductions in Hib disease have been seen in other countries
with routine childhood vaccination.5,10 Since Hib disease has become relatively
rare, cases of epiglottitis can no longer be assumed to be due to H. influenzae type
b and, moreover, even when H. influenzae is isolated from a normally sterile site,
it may not be type b. Thus, laboratory confirmation of H. influenzae infection and
serotype should always be sought before vaccination failure is assumed.11,12

132 The Australian Immunisation Handbook 9th Edition

Figure 3.4.1: Haemophilus influenzae type b (Hib) notifications, presumed Hib


hospitalisations and deaths* of children aged 0 to 4years from Hib, Australia
1993 to 20058
25

25
Deaths
Hospitalisation rate
Notification rate

20

15

15

10

10

Number of deaths

Rate per 100,000 population

20

0
1993

1994

1995

1996

1997

1998

1999

2000

2001

2002

2003

2004

2005

Year

* Hospitalisations and deaths include those for Haemophilus meningitis for the period up to
30 June 2005 (hospitalisations) and 31 December 2004 (deaths).
Notifications where themonth of diagnosis was between July 1993 and December 2005;
hospitalisations where themonth of admission was between 1 July 1993 and 30 June 2005;
deaths where the date of death was recorded between January 1993 and December 2004.

Vaccines
The first generation Hib vaccines, consisting of purified polysaccharide (PRP)
from the Hib capsule, were not effective in children <18months of age. A review
of the efficacy data for the second generation Hib vaccines, which consist of PRP
chemically linked (conjugated) to a variety of carrier proteins, found 3 of the 4
Hib vaccines to be immunogenic against invasive Hib disease, PRP-OMP, PRP-T
and HbOC.13 The fourth vaccine, PRP-D, was not found to be highly protective in
high-risk populations, such as Indigenous children.13

Haemophilus influenzae type b (Hib) 133

3.4 Haemophilus
influenzae type b (hib)

There are 2 main groups of carrier proteins associated with a different temporal
pattern of PRP antibody response. The vaccine using the outer membrane
protein of Neisseria meningitidis as a carrier protein (PRP-OMP) (COMVAX,
Liquid PedvaxHIB) gives protective PRP antibody responses after the first dose,
and requires only 2 doses to complete the primary course. For this reason, its
main application worldwide has been in populations with a high incidence
of early onset disease.5 Vaccines using other protein carriers such as tetanus
(PRP-T) (Hiberix, Infanrix hexa) and diphtheria (HbOC) toxoids do not achieve
protective PRP antibody levels until at least a second dose has been given, and

require 3 doses to complete primary immunisation. No or minimal immunologic


interference has been observed when children are vaccinated with 7vPCV and
Infanrix hexa at the same immunisation visit.14,15
Many Hib combination vaccines containing acellular pertussis are known to
produce lower Hib antibody responses than similar formulations containing
whole-cell pertussis.16 When administered according to the United Kingdoms
schedule as 3 primary doses at 2, 3 and 4months of age without a booster, their
use has been associated with an increased risk of vaccine failure.17 In other
European countries that routinely give a fourth dose around the time of the 1st
birthday, as is included in the Australian schedule, no loss of effectiveness has
been observed.18,19
Liquid PedvaxHIB CSL Biotherapies/Merck & Co Inc (PRP-OMP).
Each 0.5mL monodose vial contains 7.5g PRP conjugated to 125g
meningococcal protein; liquid formulation with 35g borax and 225g
aluminium hydroxide.
Hiberix GlaxoSmithKline (PRP-T). Each 0.5mL monodose lyophilised
vaccine contains 10g PRP conjugated to 30g tetanus toxoid (with a
lactose stabiliser) for reconstitution with 0.9% saline.
Combination vaccines that include Hib
COMVAX CSL Biotherapies/Merck & Co Inc (Hib (PRP-OMP)-hepatitis
B). Each 0.5mL monodose vial contains 7.5g PRP conjugated to 125g
meningococcal protein, 5g hepatitis B surface antigen; 225g aluminium
hydroxide; 35g borax. May contain yeast proteins.
Infanrix hexa GlaxoSmithKline (DTPa-hepB-IPV-Hib; diphtheriatetanus-acellular pertussis-hepatitis B-inactivated poliomyelitis vaccineHaemophilus influenzae type b (Hib)). The vaccine consists of both a 0.5mL
pre-filled syringe containing 30 IU diphtheria toxoid, 40 IU tetanus toxoid,
25g pertussis toxoid (PT), 25g filamentous haemagglutinin (FHA), 8g
pertactin (PRN), 10g recombinant HBsAg, 40 D-antigen units inactivated
polioviruses type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32
D-antigen units type 3 (Saukett) adsorbed onto aluminium hydroxide/
phosphate; phenoxyethanol as preservative; traces of formaldehyde,
polymyxin and neomycin and a vial containing a lyophilised pellet of 10g
purified Hib capsular polysaccharide (PRP) conjugated to 2040g tetanus
toxoid. The vaccine must be reconstituted by adding the entire contents of the
syringe to the vial and shaking until the pellet is completely dissolved. May
also contain yeast proteins.

134 The Australian Immunisation Handbook 9th Edition

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.20 Store
conjugate Hib vaccines at +2C to +8C. Do not freeze.

Dosage and administration


The dose of Hib vaccine is 0.5mL to be given by IM injection. Conjugate Hib
vaccines may be administered in separate sites on the same day as any of the
other childhood vaccines such as the 7-valent pneumococcal conjugate (7vPCV),
meningococcal serogroup C conjugate (MenCCV), hepatitis B, DTPa-containing
and monovalent IPV (or IPV-containing) vaccines.

Recommendations
(i) Hib vaccine is recommended for all infants from 2months of age
Immunisation using PRP-OMP (COMVAX or Liquid PedvaxHIB) requires 2
primary doses at 2 and 4months, followed by a booster at 12months of age. If
PRP-T (Infanrix hexa or Hiberix) is used, 3 primary doses at 2, 4 and 6months
are needed, with a booster at 12months of age.

(ii) Indigenous children living in the Northern Territory,


Queensland, South Australia and Western Australia
Many Indigenous populations experienced high Hib attack rates associated with
early peak disease onset before the introduction of Hib immunisation. While
vaccination has reduced the overall incidence of invasive Hib infection in these
vulnerable groups, increased disease risk during the first year of life remains.
It is therefore important that Aboriginal and Torres Strait Islander children in
jurisdictions (the Northern Territory, Queensland, South Australia and Western
Australia) where such different patterns of Hib disease remain evident continue
to receive PRP-OMP, because of the early antibody response seen with this
vaccine.7 In Alaskan natives, who experienced similar pre-vaccination attack rate
profiles, re-emergence of Hib disease was observed when the Hib vaccine in use
was changed from PRP-OMP to HbOC.21

(iii) Non-Indigenous children and Indigenous children living in


Australian Capital Territory, New South Wales, Tasmania and Victoria

Haemophilus influenzae type b (Hib) 135

3.4 Haemophilus
influenzae type b (hib)

Any licensed Hib vaccine may be used in these children as the period of
significant risk does not begin until after 6months of age. Although there are
limited data on the epidemiology of Hib disease before vaccination in Indigenous
children in south-eastern Australia, available data since vaccination commenced
in 1993 suggest that the epidemiology in these children does not differ
substantially from that in non-Indigenous children living in these areas (NCIRS
data, unpublished).

(iv) Interchangeability of Hib vaccines


It is recommended that the same conjugate vaccine be used for all doses.
However, if necessary, after the first dose, any Hib vaccine may be used to
complete the primary course.22 For primary vaccination, only 2 doses of PRPOMP are required, but if any other Hib vaccine is given, a total of 3 doses is
required to complete the primary course.23 This means that if the previous Hib
vaccine type is unknown for any doses, or the same vaccine type is unavailable,
the primary course can be completed with a total of 3 doses of any combination
of registered Hib vaccines. For booster doses and in children >15months of
age, regardless of previous Hib vaccinations, a single dose of any registered Hib
vaccine is sufficient for protection. Details of catch-up vaccination schedules are
given in Section 1.3.5, Catch-up.

(v) Vaccine failures


Children who have developed confirmed Hib disease after 2 or more doses
of PRP-OMP or 3 or more doses of PRP-T may warrant immunological
investigation. Consultation with an immunologist with paediatric expertise is
recommended.

(vi) Preterm babies


Preterm babies can be immunised at the normal age, without correction for
prematurity24 (see Section 2.3.2, Vaccination of women planning pregnancy, pregnant
or breastfeeding women, and preterm infants). Extremely preterm babies (<28weeks
gestation or <1500 g birth weight) who are vaccinated with PRP-OMP should be
given an extra dose at 6months of age, resulting in a 4-dose schedule at 2, 4, 6 and
12months of age.25 When other Hib vaccines, including Infanrix hexa, are used,
no change in the usual schedule is required. Preterm babies have been shown to
produce good antibody responses to all the antigens in Infanrix hexa following
administration at 2, 4 and 6months of age, although the responses to hepatitis B
and Hib are not quite as high as in term babies.

(vii) Splenectomy
Hib is an uncommon cause of post-splenectomy sepsis in adults and children.
Children >2years of age who have received all scheduled doses of Hib vaccine
do not require a booster dose after splenectomy. A single dose of Hib vaccine is
recommended for other splenectomised individuals who were not vaccinated
in infancy or are incompletely vaccinated. The vaccine should be given 2weeks
before a planned splenectomy. Subsequent booster doses of Hib vaccine are
not required.26 For other recommendations for asplenic or splenectomised
individuals, see Section 2.3.3, Vaccination of individuals with impaired immunity due
to disease or treatment.

136 The Australian Immunisation Handbook 9th Edition

(viii) Allogeneic and autologous haematopoietic


stem cell transplant (HSCT) recipients
These patients should also be considered for Hib vaccination post transplant.
The Hib conjugate vaccine should be administered to recipients at 12, 14, and
24months after HSCT. See Section 2.3.3, Vaccination of individuals with impaired
immunity due to disease or treatment.

Contraindications
The only contraindications to any of the Hib vaccines are:
anaphylaxis following a previous dose of any of the vaccines, or
anaphylaxis following any component of the vaccine.

Adverse events
Swelling and redness at the injection site after the first dose are common and
have been reported in up to 5% of vaccinated children. Fever in up to 2%
(common) has also been reported. These adverse events usually appear within
3 to 4 hours and resolve completely within 24 hours. The incidence of these
adverse events declines with subsequent doses, so it is recommended that the
course of vaccination be completed regardless.

The public health management of contacts


of a child with invasive Hib disease
Healthcare workers should be guided by public health authorities in the public
health management of cases of invasive Hib disease.

Household
As the incidence of invasive Hib disease is now very low, rifampicin chemo
prophylaxis is no longer routinely indicated unless the household contains either:
an infant <7months of age (regardless of vaccination status), or
a child aged 7months to 5years who is inadequately vaccinated according to
the Hib schedule.

Haemophilus influenzae type b (Hib) 137

3.4 Haemophilus
influenzae type b (hib)

In this case, everybody in the household should receive rifampicin prophylaxis after
a case of invasive Hib disease in any household member, with the exception of
pregnant women for whom ceftriaxone may be used. The recommended dose of
rifampicin is 20mg/kg as a single daily dose (maximum daily dose 600mg) for
4days. Neonates (<1month of age) should receive 10mg/kg daily for 4days.

Childcare facilities
Similarly, if the index case attends a child day-care facility for more than 18
hours aweek, rifampicin should be given to all children and staff who were
in the same room group (as the case) in the 7days preceding the cases onset,
provided that at least one of these close contacts is a child <24months of age who
is inadequately vaccinated. Although there may have been some intermingling
of all the children at the facility at the beginning and end of the day, this is
usually of a short duration only and not enough to justify extending the use of
rifampicin. Rifampicin prophylaxis is of no value more than 30days after initial
contact with a case.

Use in pregnancy
Refer to Chapter 2.3, Groups with special vaccination requirements, Table 2.3.1
Vaccinations in pregnancy.

Variations from product information


The product information for Hib vaccines recommends the vaccine for use in
children aged 2months to 5years. NHMRC recommends administration of Hib
vaccine to older people with asplenia or following either allogeneic or autologous
haematopoietic stem cell transplantation.
With the exception of PRP-OMP, the product information for Hib vaccines
recommends use as a booster at 18months, but the NHMRC regards a booster at
12months of age as likely to result in an equivalent immune response.
The product information for Infanrix hexa states that this vaccine may be given
as a booster dose at 18months of age. NHMRC recommends that a booster dose
of DTPa (or DTPa-containing vaccines) is not necessary at 18month of age.
However, DTPa-containing vaccine may be used for catch-up of the primary
schedule in children <8years of age.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

138 The Australian Immunisation Handbook 9th Edition

3.5Hepatitis A

Hepatitis A is an acute infection of the liver caused by a hepatovirus, the hepatitis


A virus (HAV).1 The virus survives well in the environment it persists on hands
for several hours and in food kept at room temperature for considerably longer
and is relatively resistant to heat and freezing.

Clinical features
Hepatitis A is an infection of humans; there is no animal reservoir. HAV is
predominantly transmitted by the faecal-oral route. The infecting dose is
unknown but it is presumed to be low. The incubation period of hepatitis A is
15 to 50days, with a mean of about 30days.1 HAV is excreted in faeces for up to
2weeks before the onset of illness and for at least 1week afterwards.1
In young children, HAV usually causes either an asymptomatic infection or a
very mild illness without jaundice. Patients with symptomatic illness typically
have a 4 to 10 day prodrome of systemic (fever, malaise, weakness and anorexia)
and gastrointestinal (nausea and vomiting) symptoms. Dark urine is usually
the first specific manifestation of acute hepatitis A, followed a day or 2 later
by jaundice and pale faeces. The prodromal symptoms tend to wane with the
onset of jaundice, although the anorexia and malaise may persist; pruritus and
localised hepatic discomfort or pain may follow.1 The duration of illness varies
but most patients feel better and have normal, or near normal, liver function
tests within amonth of the onset of illness. Complications of hepatitis A are
uncommon but include, on rare occasion, fulminant hepatitis.2 Hepatitis A does
not cause chronic liver disease.
The diagnosis is made by detecting anti-HAV IgM in serum during the acute
illness. Anti-HAV IgM is invariably present by the time the patient presents and
persists for 3 to 6months after the acute illness.1 Serum anti-HAV IgG indicates
past infection (or possibly immunisation) and therefore immunity; it probably
persists for life.

Epidemiology
Hepatitis A was a considerable public health problem in Australia in the
1990s. During this time numerous outbreaks occurred in child day-care centres
and preschools,3 communities of men who have sex with men,4 schools and
residential facilities for the intellectually disabled,5 and communities of injecting
drug users.4 A very large outbreak of hepatitis A associated with the consumption
of raw oysters occurred in New South Wales in 1997 (see Figure 3.5.1).6
However, there has been a marked decline in notifications of hepatitis A in
Australia in recentyears (see Figure 3.5.1). This is probably a consequence of

Hepatitis A 139

3.5 Hepatitis A

Virology

the liberal use of hepatitis A vaccine among travellers, and those at increased
risk because of lifestyle or occupation. A hepatitis A vaccination program for
Indigenous children in north Queensland that began in 1999 has also contributed
substantially to the decline in notifications.7
Nevertheless, Indigenous Australian children remain at considerably greater
risk, not only of acquiring hepatitis A but also for being hospitalised with the
infection, compared to non-Indigenous children.8 This is particularly true for
Indigenous children residing in other regions of Queensland, the Northern
Territory, South Australia and Western Australia.
Figure 3.5.1: Notifications of hepatitis A in Australia, 1991 to 2006


"OPVUCSFBLBTTPDJBUFEXJUIUIF
DPOTVNQUJPOPGPZTUFSTJO/48



/PUJGJDBUJPOT





*OUSPEVDUJPOPG
WBDDJOBUJPOGPS
*OEJHFOPVTDIJMESFOJO
/PSUI2ME




+BO
+VM
+BO
+VM
+BO
+VM
+BO
+VM
+BO
+VM
+BO
+VM
+BO
+VM
+BO
+VM
+BO
+VM
+BO
+VM
+BO
+VM
+BO
+VM
+BO
+VM
+BO
+VM
+BO
+VM
+BO
+VM

.POUIBOEZFBS

Vaccines
Avaxim Sanofi Pasteur Pty Ltd (formaldehyde inactivated hepatitis A
virus (GBM strain)). Each 0.5mL pre-filled syringe contains 160 ELISA units
of hepatitis A virus (HAV) antigens inactivated by formaldehyde; 0.3mg
aluminium hydroxide; 2.5 L phenoxyethanol; 12.5g formaldehyde; trace
of neomycin.
Havrix Junior GlaxoSmithKline (formaldehyde inactivated hepatitis A
virus (HM175 strain)). Each 0.5mL monodose vial or pre-filled syringe
contains 720 ELISA units of HAV antigens; 0.25mg as aluminium
hydroxide; 0.5% w/v phenoxyethanol; traces of formaldehyde and
neomycin.
Havrix 1440 GlaxoSmithKline (formaldehyde inactivated hepatitis A virus
(HM175 strain)). Each 1.0mL monodose vial or pre-filled syringe contains
1440 ELISA units of HAV antigens; 0.5mg aluminium hydroxide; 0.5% w/v
phenoxyethanol; traces of formaldehyde and neomycin.

140 The Australian Immunisation Handbook 9th Edition

Twinrix (720/20) GlaxoSmithKline (formaldehyde inactivated hepatitis


A virus (HM175 strain) and recombinant hepatitis B vaccine). Each 1.0mL
monodose vial or syringe contains 720 ELISA units of HAV antigens, 20g
recombinant DNA hepatitis B surface antigen protein; 0.45mg aluminium
phosphate/hydroxide; 0.5% w/v phenoxyethanol; traces of formaldehyde
and neomycin. May contain yeast proteins.
VAQTA Paediatric/Adolescent formulation CSL Biotherapies/Merck &
Co Inc (formaldehyde inactivated hepatitis A virus (CR326F strain)). Each
0.5mL monodose vial contains approximately 25 units (U) of hepatitis
A virus protein; 0.225mg aluminium hydroxide; 35g borax; trace of
formaldehyde.
VAQTA Adult formulation CSL Biotherapies/Merck & Co Inc
(formaldehyde inactivated hepatitis A virus (CR326F strain)). Each 1.0mL
monodose vial contains approximately 50 units (U) of hepatitis A virus
protein; aluminium 0.45mg as aluminium hydroxide; 70g borax; trace of
formaldehyde.
Vivaxim Sanofi Pasteur Pty Ltd (inactivated hepatitis A virus and typhoid
Vi capsular polysaccharide). Supplied in a unique dual-chamber syringe
which enables the 2 vaccines to be mixed just before administration. Each
1.0mL dose of mixed vaccine contains 160 ELISA units of inactivated
hepatitis A virus antigens, 25g purified typhoid capsular polysaccharide;
0.3mg aluminium hydroxide; 2.5 L phenoxyethanol; formaldehyde; traces
of neomycin and bovine serum albumin.
The inactivated hepatitis A vaccines are prepared from HAV harvested from
human diploid cell cultures, which are then purified by ultrafiltration and
chromatography, inactivated by formaldehyde, and then adsorbed onto
aluminium hydroxide adjuvant. Although the vaccines are prepared from
differing strains of HAV, there is only one known serotype; immunity induced by
a particular strain probably provides protection against all strains.1
The Avaxim, Havrix, Twinrix and Vivaxim vaccines contain a preservative,
2-phenoxyethanol. All the vaccines contain minute amounts of residual
formaldehyde. Although the manufacturers use slightly different production
methods and quantify the HAV antigen content in their respective vaccines

Hepatitis A 141

3.5 Hepatitis A

Twinrix Junior (360/10) GlaxoSmithKline (formaldehyde inactivated


hepatitis A virus (HM175 strain) and recombinant hepatitis B vaccine). Each
0.5mL monodose vial or pre-filled syringe contains 360 ELISA units of
HAV antigens, 10g recombinant DNA hepatitis B surface antigen protein;
0.225mg aluminium phosphate/hydroxide; 0.5% w/v phenoxyethanol;
traces of formaldehyde and neomycin. May contain yeast proteins.

differently, the equivalent vaccines of the different manufacturers are


interchangeable.
The inactivated hepatitis A vaccines induce HAV antibodies (anti-HAV) at titres
many-fold greater than that provided by the recommended dose of normal
human immunoglobulin. Although the vaccines are highly immunogenic (see
below), the titres are usually below the detection limits of the routinely available
commercial tests for anti-HAV.1 Therefore, serological testing to assess immunity
after vaccination against hepatitis A is neither necessary nor appropriate. Likewise, it
is also inappropriate to undertake testing if an individual cannot recall if he/she
has been vaccinated against hepatitis A in the past; if no vaccination records are
available, vaccination should be advised.
Hepatitis A vaccines are highly immunogenic in both children and adults, with
virtually universal seroconversion 4weeks after vaccination.1 Two randomised
clinical trials conducted in the early 1990s showed that the vaccines have a very
high protective efficacy, approaching 100%.9,10 This finding is supported by
the apparent eradication of hepatitis A from Indigenous communities in north
Queensland since the introduction of the vaccination program in the region.7
The duration of immunity and, therefore, protection following vaccination is not
certain. However, vaccine-induced anti-HAV probably persists for manyyears.
There is no current evidence that booster doses are required; in healthy
individuals, it is quite possible that they will never be required.11

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.12 Hepatitis
A vaccines should be transported and stored at +2C to +8C. Do not freeze.

Dosage and administration


The inactivated hepatitis A vaccines are administered by IM injection. The
recommended dosages and schedules for use in Australia are given in Table 3.5.1.

142 The Australian Immunisation Handbook 9th Edition

Table 3.5.1: Recommended dosages and schedules for use of the inactivated
hepatitis A vaccines
Vaccinees age
(years)

Dose
Volume
(HAV antigen) per dose
(mL)

Vaccination schedule
(mo=months)

Monovalent hepatitis A vaccines


Avaxim

160 EIA U

0.5

0, 6 to 12 mo

Havrix Junior

2 <16

720 EIA U

0.5

0, 6 to 12 mo

Havrix 1440

16

1440 EIA U

1.0

0, 6 to 12 mo

VAQTA Paediatric/ 1 <18


Adolescent

25 U

0.5

0, 6 to 18 mo

VAQTA Adult

50 U

1.0

0, 6 to 18 mo

18

Combination hepatitis A/hepatitis B vaccines


Twinrix Junior
(360/10)

1 <16

360 EIA U

0.5

0, 1, 6 mo

Twinrix (720/20)

16

720 EIA U

1.0

0, 1, 6 mo

Twinrix (720/20)*

1 <16

720 EIA U

1.0

0, 6 to 12 mo

Twinrix (720/20)

16

720 EIA U

1.0

0, 7, 21days, 12 mo

1.0 (mixed
vaccine)

0; a single dose of
monovalent adult
formulation hepatitis
A vaccine should be
given at 6 to 36 mo.

Combination hepatitis A/typhoid vaccine


Vivaxim

16

160 EIA U

* This schedule should not be used for those who require prompt protection against
hepatitis B; for example, if there is close contact with a known hepatitis B carrier.
This rapid schedule should be used only if there is very limited time before departure to
either moderately or highly endemic regions.

Recommendations
To avoid unnecessary vaccination, it is recommended that the following groups
be screened for pre-existing natural immunity to hepatitis A:
those born before 1950,
those who spent their early childhood in endemic areas, and
those with an unexplained previous episode of hepatitis or jaundice. (NB.
Such a previous episode cannot be assumed to be hepatitis A.)
If, upon screening, a person has either total hepatitis A antibodies or anti-HAV
IgG, he/she has presumably had previous, perhaps unrecognised, HAV infection
(or less likely, has been previously immunised) and can be assumed to be
immune and, therefore, does not need hepatitis A vaccination.

Hepatitis A 143

3.5 Hepatitis A

Vaccine

(i) Hepatitis A vaccination is recommended for:


all travellers to, and all expatriates living in, moderately to highly endemic
areas (including all developing countries)
A single dose of a monovalent hepatitis A vaccine provides protective levels of
anti-HAV for at least a year;1 the second dose is recommended to increase the
duration of protection. As they do not contain live viruses, hepatitis A vaccines
can be administered either simultaneously with, or within amonth of, all other
vaccines relevant to international travel.13
There is no place for the routine use of normal human immunoglobulin to
prevent hepatitis A in travellers. It should only be given (at the same time
as hepatitis A vaccine) to those, such as aid-workers about to be deployed in
emergency refugee camps, who will be living in very inadequate circumstances.
Otherwise, it is only recommended for contacts of hepatitis A cases (see The
public health management of contacts of hepatitis A cases below).
Aboriginal and Torres Strait Islander children residing in the Northern
Territory, Queensland, South Australia and Western Australia
Hepatitis A vaccination for these children should commence in the second year
of life. State/Territory health authorities should be contacted about the local
hepatitis A vaccination schedules, including catch-up.
those whose occupation may put them at risk of acquiring hepatitis A
This includes those who live or work in rural and remote Indigenous
communities, child day-care and preschool personnel, carers of people with
intellectual disabilities, healthcare workers who regularly provide care for
Aboriginal and Torres Strait Islander children, plumbers or sewage workers, and
sex workers.
those whose lifestyle may put them at risk of acquiring hepatitis A
This includes men who have sex with men, and injecting drug users.
people with intellectual disabilities
people chronically infected with either hepatitis B or hepatitis C viruses
patients with chronic liver disease
Hepatitis A vaccination is recommended for patients with chronic liver disease
of any aetiology. Those with chronic liver disease of mild to moderate severity
mount a satisfactory immune response following vaccination, but those with
end-stage liver disease do not respond as well, and liver transplant recipients
may not respond at all.14,15 Nevertheless, all those with chronic liver disease
should be vaccinated, preferably as early in the course of the disease as possible.

144 The Australian Immunisation Handbook 9th Edition

(ii) Combined hepatitis A/hepatitis B vaccines


Combined hepatitis A/hepatitis B vaccines should be considered for:
NB. Twinrix (720/20) can be administered according to a rapid schedule if
there is limited time before departure.16 This consists of a single dose on each
ofdays 0, 7 and 21. It is important that a fourth dose be given as a booster
12months after the first dose to ensure longer-term protection.
medical, dental and nursing undergraduate students,
men who have sex with men,
sex industry workers,
injecting drug users,
patients with chronic liver disease,
solid organ transplant recipients (see Table 2.3.2 Recommendations for
vaccinations for solid organ transplant (SOT) recipients),
people with intellectual disabilities and their carers.
NB. Twinrix (720/20) can be administered in a 2-dose regimen in people 1 to
15years of age (see Table 3.5.1). However, this regimen should not be used in
those who require prompt protection against hepatitis B; for example, if there is
close contact with a known hepatitis B carrier.
Combined hepatitis A/hepatitis B vaccines can be administered simultaneously
with, or within amonth of, all other vaccines relevant to international travel.

(iii) Combined hepatitis A/typhoid vaccine


The combined hepatitis A/typhoid vaccine can be recommended for all
those 16years of age who intend travelling to developing countries, and is
particularly useful for those already immunised against hepatitis B. The vaccine
can be administered simultaneously with, or within amonth of, all other vaccines
relevant to international travel.
A single dose of a monovalent adult formulation hepatitis A vaccine 6 to
36months later is required to provide longer-term protection against hepatitis
A. A booster dose of typhoid capsular polysaccharide vaccine is required after
3years if there is a continued risk. The combined hepatitis A/typhoid vaccine
may be used as a booster vaccine if a person received a previous dose of a
monovalent adult formulation hepatitis A vaccine. This may be given 6 to
36months after primary vaccination.

Hepatitis A 145

3.5 Hepatitis A

expatriates and long-term visitors to developing countries,

Contraindications
The only contraindications to any of the hepatitis A vaccines are:
anaphylaxis following a previous dose of any of the hepatitis A vaccines, or
anaphylaxis following any component of the vaccine.
Combination vaccines containing the hepatitis B component are contraindicated
where there is a history of anaphylaxis to yeast.

Adverse events
The most common adverse events following administration of hepatitis A
vaccines are mild local events of a short duration, probably caused by the
aluminium hydroxide adjuvant. About 15% (very common) of adults report
headache and approximately 5% (common) report malaise or fatigue following
vaccination.17 Up to 20% (very common) of children who received either Havrix
or VAQTA experienced soreness at the injection site. In both adults and children,
systemic adverse events such as headache and fever are much less common than
local adverse events.17
Hepatitis A vaccines do not affect liver enzyme levels. They can be safely given to
HIV-infected people, and do not adversely affect either the HIV load or CD4 cell
count.18

The public health management of


contacts of hepatitis A cases
Normal human immunoglobulin (NHIG) can be used to prevent secondary cases
in close contacts of hepatitis A cases. (NB. A hepatitis A IgM positive test in an
adult without either clinical or epidemiological features of hepatitis A should be
considered as a false-positive result.19 In this circumstance, no interventions are
necessary for the close contacts.)
NHIG should be administered to close contacts within 2weeks (of the last
exposure to the cases) in the doses given in Table 3.5.2; NHIG may not be
effective if given >2weeks after the exposure.17 Close contacts are those who
have had contact with a case during the 2weeks before, up until 1week after, the
onset of jaundice, and usually include only household and/or sexual contacts
(but in some circumstances may include close occupational exposure).
Table 3.5.2: Recommended doses of normal human immunoglobulin (NHIG) to
be given as a single intramuscular injection to close contacts of hepatitis A cases
Weight

Dose NHIG

Under 25kg

0.5mL

2550kg

1.0mL

Over 50kg

2.0mL

146 The Australian Immunisation Handbook 9th Edition

Although 1 study suggests that hepatitis A vaccine may be effective in preventing


secondary cases of hepatitis A in close contacts,20 there is currently insufficient
evidence to be able to recommend it for this purpose.
If a person with hepatitis A was a food-handler by occupation while infectious,
a review of the food-handling procedures in the food establishment should
be undertaken and the staff at the establishment reminded of standard food
and personal hygiene practices.17 If the review identifies issues which raise the
possibility of transmission of HAV, NHIG should be administered to the other
food-handlers in the establishment. State/Territory public health authorities
should determine the need for recall of customers of the establishment for NHIG.
A food-handler with hepatitis A should be excluded from work until at least
1week after the onset of jaundice.
A single case of hepatitis A associated with a day-care or preschool facility (ie. a
case in an attendee child, a staff member or a household contact of an attendee or
staff member) does not require any mass intervention.3 However, the supervisor
of the facility should be contacted to:
explore the possibility of within-centre transmission by the case (eg. faecal
accidents, other hygiene control concerns), and
determine if there could be other cases associated with the facility. In
particular, it should be ascertained whether an attendee child arrived from an
endemic region overseas about amonth before the cases onset, and whether
any other children at the facility have recently been vaguely unwell with a
change in bowel motions.
Should there be any concerns about the potential for further transmission of
hepatitis A virus within the facility, mass interventions (as per 2 or more cases
below) may be considered. The supervisor should be reminded of the relevant
infection control practices that should be in place at all times at the facility,
and that hepatitis A vaccine is routinely recommended for day-care and
preschool staff (unless they have either had hepatitis A in the past or been
vaccinated previously). A useful reference is Staying Healthy in Child Care
available at http://www.nhmrc.gov.au/publications/synopses/ch43syn.htm.
Two or more cases of hepatitis A (associated with the same day-care or
preschool facility) that occur in different households are strongly suggestive that
transmission of HAV is occurring within that facility.3 These cases may be in
attendee children or staff or household contacts of an attendee or staff member.
As soon as transmission of HAV is recognised within a day-care or preschool
facility, NHIG should be offered to children and susceptible staff in the relevant
age groups (or classes) at that facility. Parents and staff need to be reminded
that live virus vaccines, MMR and varicella vaccines in particular, should not be
administered within 3months of receiving IM NHIG.3

Hepatitis A 147

3.5 Hepatitis A

Further public health considerations

Use in pregnancy
Refer to Chapter 2.3, Groups with special vaccination requirements, Table 2.3.1
Vaccinations in pregnancy.

Variations from product information


None.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

148 The Australian Immunisation Handbook 9th Edition

3.6Hepatitis B
Virology
Hepatitis B virus (HBV) contains partially double-stranded DNA. The outer
surface of the virus is glycolipid which contains the hepatitis B surface antigen
(HBsAg). Other important antigenic components are hepatitis B core antigen
(HBcAg), and hepatitis B e antigen (HBeAg). HBcAg is not detectable in serum,
but can be detected in liver tissue in people with acute or chronic hepatitis B
infection. Antibodies developed to HBsAg (anti-HBs) indicate immunity, whereas
persistence of HBsAg denotes infectivity, which is greater if HBeAg and HBV
DNA are positive.1

In approximately 30 to 50% of adults, infection causes symptomatic acute


hepatitis, but in young children, particularly those <1 year of age, infection is
usually asymptomatic. The incubation period is 45 to 180days and the period
of communicability extends from severalweeks before the onset of acute illness
usually to the end of the period of acute illness. Acute illness is indistinguishable
from other forms of hepatitis, and symptoms include fever, jaundice, malaise,
anorexia, nausea and vomiting, abdominal pain (especially in the right upper
quadrant), myalgia, and the passage of dark-coloured urine and light-coloured
stools. Jaundice may be preceded by an acute febrile illness with arthralgia or
arthritis and rash, most typical of hepatitis B. During recovery, malaise and
fatigue may persist for manyweeks. Fulminant hepatitis occurs in approximately
1% of acute cases.1,2
Following acute infection, 1 to 10% of those infected as adults2,3 and up to 90%
of those infected as neonates1,2 remain persistently infected for manyyears (see
Figure 3.6.1). Chronically infected carriers of HBV are identified by the long-term
presence (longer than 6months) of circulating HBsAg.4

Hepatitis B 149

3.6 Hepatitis B

Clinical features

Figure 3.6.1: The influence of age of infection with the hepatitis B virus on the
likelihood of becoming a hepatitis B carrier

(Modified and used with permission from: Edmunds WJ, Medley GF, Nokes DJ, Hall AJ, Whittle HC. The
influence of age on the development of the hepatitis B carrier state. Proceedings. The Royal Society Biological
Sciences.1993;253:197-201.)

Carriers of HBV are capable of transmitting the disease, though often remain
asymptomatic and may not be aware that they are infected. Most of the serious
complications associated with hepatitis B infection occur in HBV carriers.
Chronic active hepatitis develops in more than 25% of carriers, and up to 25% die
prematurely of cirrhosis or hepatocellular carcinoma.1,2

Epidemiology
The prevalence of HBV carriage differs in different parts of the world, and may
be quite variable within countries. Carrier rates vary from 0.1 to 0.2% among
Caucasians in the United States, northern Europe and Australia, 1 to 5% in
the Mediterranean countries, parts of eastern Europe, China, Africa, Central
and South America, and some Australian Aboriginal populations, and greater
than 10% in many sub-Saharan African, southeast Asian and Pacific island
populations.5-7 First-generation immigrants usually retain the carrier rate of
their country of origin, but subsequent generations show a declining carrier rate
irrespective of vaccination.5
Transmission of hepatitis B may result from percutaneous inoculation or mucosal
contact with blood or sexual secretions from an HBsAg-positive individual.
Screening of blood and organ donors has virtually eliminated the risk of
transmission of hepatitis B through blood transfusion and organ transplants.8,9
Saliva may also contain levels of virus which are likely to be infective only if
inoculated directly into tissue (ocular or mucous membranes). Transmission by
inadvertent parenteral inoculation, such as by toothbrush, razor etc., through
close personal contact in households in which 1 or more carriers or other infected
individuals reside, is a low but significant risk.
Routes of transmission include:
sharing injecting equipment (such as occurs in injecting drug use),
needle-stick injury, and other types of parenteral inoculation,

150 The Australian Immunisation Handbook 9th Edition

sexual contact (including heterosexual or homosexual intercourse, although


the latter has a higher risk),
transmission from infected mother to neonate (vertical transmission), usually
occurring at or around the time of birth,
child-to-child (horizontal) transmission, usually through contact between open
sores or wounds,
breastfeeding,10
nosocomial transmission in overseas healthcare facilities if infection control
procedures are unsatisfactory.

Australian vaccination policy

Vaccines
Engerix-B GlaxoSmithKline (recombinant DNA hepatitis B vaccine).
Adult formulation Each 1.0mL monodose vial contains 20g
recombinant hepatitis B surface antigen (HBsAg) protein, adsorbed onto
0.5mg aluminium hydroxide. Paediatric formulation Each 0.5mL
monodose vial contains 10g HBsAg protein, adsorbed onto 0.25mg
aluminium hydroxide. Both formulations contain traces of yeast proteins
and thiomersal (<2g/mL). Both are available in packs of 10.
H-B-VAX II CSL Biotherapies/Merck & Co Inc (recombinant DNA hepatitis
B vaccine). Adult formulation preservative free Each 1.0mL pre-filled
syringe or vial contains 10g recombinant HBsAg protein, adsorbed onto
0.5mg aluminium hydroxide. May contain yeast proteins. Paediatric
formulation preservative free Each 0.5mL pre-filled syringe or vial
contains 5g recombinant HBsAg protein, adsorbed onto 0.25mg aluminium
hydroxide. May contain yeast proteins. Both are available in packs of 10.
Dialysis formulation preservative free Each 1.0mL vial contains 40g
recombinant HBsAg protein, adsorbed onto 0.5mg aluminium hydroxide.
May contain yeast proteins. Available as single pack only.

Hepatitis B 151

3.6 Hepatitis B

The initial strategy for the control of hepatitis B in Australia commenced in


1988, targeting groups at particular risk of infection for vaccination at birth. In
addition to vaccine, hepatitis B immunoglobulin (HBIG) was given if the mother
was a hepatitis B carrier. In 1990, universal infant vaccination commenced
in the Northern Territory. In 1996, the NHMRC recommended a universal
hepatitis B vaccination program for infants and adolescents. The adolescent
program commenced in some States and Territories in 1997 and the universal
infant program, with the first dose given at birth, began nationally in 2000. The
adolescent program will continue until those immunised for hepatitis B in the
childhood program reach adolescence.

Combination vaccines that include both DTPa and hepatitis B


Infanrix hexa GlaxoSmithKline (DTPa-hepB-IPV-Hib; diphtheriatetanus-acellular pertussis-hepatitis B-inactivated poliomyelitis vaccineHaemophilus influenzae type b (Hib)). The vaccine consists of both a 0.5mL
pre-filled syringe containing 30 IU diphtheria toxoid, 40 IU tetanus toxoid,
25g pertussis toxoid (PT), 25g filamentous haemagglutinin (FHA), 8g
pertactin (PRN), 10g recombinant HBsAg, 40 D-antigen units inactivated
polioviruses type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32
D-antigen units type 3 (Saukett) adsorbed onto aluminium hydroxide/
phosphate; phenoxyethanol as preservative; traces of formaldehyde,
polymyxin and neomycin and a vial containing a lyophilised pellet of 10g
purified Hib capsular polysaccharide (PRP) conjugated to 2040g tetanus
toxoid. The vaccine must be reconstituted by adding the entire contents of the
syringe to the vial and shaking until the pellet is completely dissolved. May
also contain yeast proteins.
Infanrix Penta GlaxoSmithKline (DTPa-hepB-IPV; diphtheria-tetanusacellular pertussis-hepatitis B-inactivated poliomyelitis vaccine). Each
0.5mL pre-filled syringe contains 30 IU diphtheria toxoid, 40 IU tetanus
toxoid, 25g PT, 25g FHA, 8g PRN, 10g recombinant HBsAg, 40
D-antigen units inactivated polioviruses type 1 (Mahoney), 8 D-antigen
units type 2 (MEF-1) and 32 D-antigen units type 3 (Saukett) adsorbed onto
aluminium hydroxide/phosphate; phenoxyethanol as preservative; traces of
formaldehyde, polymyxin and neomycin. May also contain yeast proteins.
Other combination vaccines that include hepatitis B
COMVAX CSL Biotherapies/Merck & Co Inc (Hib (PRP-OMP)-hepatitis
B). Each 0.5mL monodose vial contains 7.5g PRP conjugated to 125g
meningococcal protein, 5g hepatitis B surface antigen; 225g aluminium
hydroxide; 35g borax. May contain yeast proteins.
Twinrix Junior (360/10) GlaxoSmithKline (formaldehyde inactivated
hepatitis A virus (HM175 strain) and recombinant hepatitis B vaccine). Each
0.5mL monodose vial or pre-filled syringe contains 360 ELISA units of
HAV antigens, 10g recombinant DNA hepatitis B surface antigen protein;
0.225mg aluminium phosphate/hydroxide; 0.5% w/v phenoxyethanol;
traces of formaldehyde and neomycin. May contain yeast proteins.
Twinrix (720/20) GlaxoSmithKline (formaldehyde inactivated hepatitis
A virus (HM175 strain) and recombinant hepatitis B vaccine). Each 1.0mL
monodose vial or syringe contains 720 ELISA units of HAV antigens, 20g
recombinant DNA hepatitis B surface antigen protein; 0.45mg aluminium
phosphate/hydroxide; 0.5% w/v phenoxyethanol; traces of formaldehyde
and neomycin. May contain yeast proteins.

152 The Australian Immunisation Handbook 9th Edition

Hepatitis B vaccines are prepared using recombinant technology. After


purification, the HBsAg protein is adsorbed onto elemental aluminium (as
hydroxide and/or phosphate). Preservatives, including thiomersal, may be
added. Hepatitis B vaccines may contain up to 1% yeast proteins (but no yeast
DNA).
Thiomersal-free vaccines, such as H-B-VAX II preservative free paediatric
formulation, are now available and are recommended for administration to
newborns and infants.11 Engerix-B paediatric formulation contains a trace amount
of thiomersal (<2g/mL). All other infant and childhood hepatitis B-containing
combination vaccines, such as Infanrix Penta, Infanrix hexa, COMVAX and
Twinrix Junior (360/10), are thiomersal-free.

Transport, storage and handling

Dosage and administration


Monovalent hepatitis B vaccines are white, slightly opalescent liquids. Any
visible change in the product, such as an amorphous flocculent or a granular
precipitate may indicate incorrect storage conditions.
(i) Administer by deep IM injection.
(ii) 3-dose regimen
For children and young adults <20years of age, a total of 3 doses of 0.5mL of
paediatric formulation is recommended. The optimal interval is 1month between
the first and second doses and a third dose 5months after the second dose. The
use of longer time intervals between doses does not impair the immunogenicity
of hepatitis B vaccine, especially in adolescents and young children.13,14 The
minimum interval between the second and third doses is 2months.
(iii) For adults 20years of age, a full course of hepatitis B vaccine consists of 3
doses of 1mL of adult formulation. There should be an interval of 1 to 2months
between the first and second doses with a third dose 2 to 5months after the
second dose (this schedule applies to both Engerix-B and H-B-VAX II). The
minimum interval between the second and third doses is 2months.
This induces protective levels of neutralising antibody against hepatitis B
virus in more than 90% of adults. The frequency of seroconversion increases
progressively from approximately 35% after the first injection to more than
90% after the third injection. There is evidence of immunity (anti-HBs) in most
vaccinated subjects after administration of 2 doses of the 3-dose vaccine regimen.
However, the third dose is necessary to increase the percentage of responders
and to provide long-term protection.

Hepatitis B 153

3.6 Hepatitis B

Transport according to National Vaccine Storage Guidelines: Strive for 5.12 Store at
+2C to +8C. Do not freeze.

(iv) Alternative 2-dose regimens


A randomised controlled trial, involving 1026 adolescents, demonstrated that
adolescents 1115years of age who received 2 doses of the adult formulation at 0
and 46months, developed similar protective antibody levels to those vaccinated
using the paediatric formulations in the standard 3-dose regimen administered at
0, 1 and 612months.15
An open label comparative study in adults found increased compliance among
those receiving a 2-dose schedule (86%) over those who completed the 3-dose
schedule (18%). Antibody responses were found to be similar among the 2
groups.16
A 2-dose schedule used in the 1115years age group will improve compliance
and provide comparable immunogenicity to that of a 3-dose paediatric schedule.
Adolescents (1115years of age) can be vaccinated with H-B-VAX II 10g (adult
formulation) or Engerix-B 20g (adult formulation) in a 2-dose regimen of 0 and
46months (H-B-VAX II) or 0 and 6months (Engerix-B). In older adolescents
up to the age of 19 years, in whom compliance with a 3-dose paediatric dosing
schedule is in doubt, a 2-dose schedule using an adult formulation may also be
used in order to improve protection.
When protection is required against both hepatitis A and hepatitis B in children
115years of age, administration of Twinrix (720/20) in a 2-dose regimen at 0
and 612months results in protective antibody levels for both hepatitis A and
hepatitis B (see Table 3.6.1).

154 The Australian Immunisation Handbook 9th Edition

Table 3.6.1: Hepatitis B and hepatitis A/hepatitis B combination vaccination


schedules
Vaccine

Age

Dose (HBsAg
protein)

Volume

Schedule (mo=months)

Monovalent hepatitis B vaccines


<20years

10g

0.5mL

0, 1, 6 mo (3-dose
schedule)

Engerix-B (adult)

1115years

20g

1.0mL

0, 6 mo (2-dose schedule)

Engerix-B (adult)

20years

20g

1.0mL

0, 1, 6 mo (3-dose
schedule)

H-B-VAX II
(paediatric)

<20years

5g

0.5mL

0, 1, 6 mo (3-dose
schedule)

H-B-VAX II (adult) 1115years

10g

1.0mL

0, 46 mo (2-dose
schedule)

H-B-VAX II (adult) 20years

10g

1.0mL

0, 1, 6 mo (3-dose
schedule)

H-B-VAX
II (dialysis
formulation)

40g

1.0mL

0, 1, 6 mo (3-dose
schedule)

20years

Combination hepatitis A/B vaccines


Twinrix (720/20)*

1 <16years

20g

1.0mL

0, 612 mo (2-dose
schedule)

Twinrix Junior
(360/10)

1 <16years

10g

0.5mL

0, 1, 6 mo (3-dose
schedule)

Twinrix (720/20)

16years

20g

1.0mL

0, 1, 6 mo (3-dose
schedule)

* This schedule should not be used for those who require prompt protection against
hepatitis B; for example, if there is close contact with a known hepatitis B carrier.

Hepatitis B 155

3.6 Hepatitis B

Engerix-B
(paediatric)

(v) Accelerated schedule


Engerix-B formulations (paediatric and adult) and Twinrix (720/20) are
registered for use in accelerated schedules. Accelerated schedules should only
be used if there is very limited time before departure to endemic regions (see
Table3.6.2).
Table 3.6.2: Accelerated hepatitis B vaccination schedules*
Vaccine

Age

Dose (HBsAg
protein)

Volume

Schedule (mo=months)

Engerix-B
(paediatric)

<20years

10g

0.5mL

0, 1, 2, 12 mo

Engerix-B (adult)

20years

20g

1.0mL

0, 1, 2, 12 mo
or
0, 7, 21days, 12 mo

Twinrix (720/20)

16years

20g

1.0mL

0, 7, 21days, 12 mo

* As higher seroprotective rates are seen after the 0, 1, 2month schedule, it is recommended
that the 0, 7, 21days schedule be used only in adults and only in exceptional circumstances.
In both schedules, a booster dose at 12months is recommended for long-term protection.

Recommendations
(i) Infants and young children
A birth dose of thiomersal-free monovalent hepatitis B vaccine, followed by
doses given in combination vaccines (such as DTPa-hepB, DTPa-hepB-IPV,
DTPa-hepB-IPV-Hib or Hib (PRP-OMP)-hepB) at 2, 4 and either 6 or 12months,
is recommended for all children.
The rationale for the universal birth dose is not only to prevent vertical
transmission from a carrier mother (recognising that there may be errors or
delays in maternal testing, reporting, communication or appropriate response),
but also to prevent horizontal transmission in the firstmonths of life from a
carrier among household or other close contacts.17 The birth dose should be given
as soon as the baby is physiologically stable, and preferably within 24 hours of
birth. Every effort should be made to administer the vaccine before discharge
from the obstetric hospital.
Extensive experience indicates that the birth dose of hepatitis B vaccine is
very well tolerated by newborn infants. It does not interfere with either the
establishment or maintenance of breastfeeding, and it is not associated with
an increased risk of either fever or medical investigation for sepsis in the
newborn.18-20
If an infant has missed the birth dose and is aged 8days or older, a catchup schedule is not required. A primary course of a hepatitis B-containing

156 The Australian Immunisation Handbook 9th Edition

combination vaccine should be given at 2, 4 and either 6 or 12months of age


(provided the mother is HBsAg negative).
NB. All babies (preterm or term) of carrier mothers must be given a birth dose of
hepatitis B vaccine and HBIG.

Management of infants born to hepatitis B carrier mothers

The first dose of monovalent hepatitis B vaccine should be given at the same
time as HBIG, but in the opposite anterolateral thigh, as soon as possible
preferably within 24 hours of birth, and definitely within 7days. This regimen
results in seroconversion rates of more than 90% in neonates, despite concurrent
administration of HBIG. If concurrent administration is not possible, vaccination
should not be delayed beyond 7days after birth as (providing it is given early)
vaccine alone has been shown to be effective in preventing carriage.21 Three
subsequent doses of a multivalent/combination vaccine should be given at 2,
4 and either 6 or 12months of age (depending on the vaccine used), so that the
infant is given a total of 4 doses of hepatitis B-containing vaccines.

Preterm babies
Preterm babies do not respond as well to hepatitis B-containing vaccines as term
babies.22-25 Thus, for babies at <32weeks gestation or <2000 g birth weight, it is
recommended to give vaccine at 0, 2, 4 and 6months of age and either:
(a) measure anti-HBs at 7months of age and give a booster at 12months of age if
antibody titre is <10mIU/mL, or
(b) give a booster at 12months of age without measuring the antibody titre.

(ii) Adolescents
Vaccination of adolescents 10 to 13years of age is recommended for all those
in this age group who have not already received a primary course of hepatitis
B vaccine. Please refer to your State/Territory health authority for further
information (see Appendix 1, Contact details for Australian, State and Territory
Government health authorities and communicable disease control).

Hepatitis B 157

3.6 Hepatitis B

Routine antenatal screening for HBsAg is essential for correct implementation


of the strategy to prevent newborn infants from becoming infected with, and
therefore carriers of, HBV. It also has benefits of enabling appropriate followup and management of a carrier, identification of the immune status of other
household members, and protection of those who are susceptible to HBV
infection. Infants born to HBsAg positive mothers should be given HBIG and
a dose of thiomersal-free monovalent hepatitis B vaccine on the day of birth.
The dose of HBIG is 100 IU to be given by IM injection. Administration of
HBIG is preferable within 12 hours of birth, as its efficacy decreases markedly if
administration is delayed beyond 48 hours after birth.

(iii) Adults for whom hepatitis B vaccination is recommended


Note: the combined hepatitis A/hepatitis B vaccine should be considered for susceptible
individuals in the groups marked with an asterisk (*).
Household contacts of acute and chronic hepatitis B carriers
There is a low, but definite, risk of transmission from a person with acute or
chronic hepatitisB. This can be reduced by avoiding contact with blood or other
body fluids and not sharing household items which can penetrate skin (such as
combs, nail brushes, toothbrushes and razors).
The risk of contacts acquiring hepatitis B infection varies according to the HBeAg
status of the carrier, and with cultural and socioeconomic factors. However, it
should be recognised that in many situations, family members may have been
exposed by the time the risk is recognised. Testing before planned vaccination
is recommended for such families, as well as for members of families who have
migrated from high prevalence countries.
Sexual contacts
Susceptible (anti-HBc and anti-HBs negative) sexual partners of patients
with acute hepatitis B should be offered post-exposure HBIG and hepatitis B
vaccination; both should be initiated within 14days of the last sexual contact.
Susceptible partners of asymptomatic carriers should also be offered vaccination.
Hepatitis B is relatively common in clients of sexual health services and
vaccination should be offered to susceptible individuals at the time of first
attendance.
*Sexually active men who have sex with men should be vaccinated, unless
they are already HBsAg positive or have serological evidence of immunity. The
combined hepatitis A/hepatitis B vaccine may be appropriate for men who have
sex with men, if they are not immune to either disease, as they are at increased
risk of both.
Haemodialysis patients, HIV-positive individuals and other adults with
impaired immunity
Dialysis patients, HIV-positive individuals and other adults with impaired
immunity should be given a larger than usual dose of hepatitis B vaccine. Adults
should be given either (i) 1mL of normal adult formulation in each arm on each
occasion (double dose), or (ii) a single dose of dialysis formulation vaccine on
each occasion, at 0, 1 and 6months. HIV-positive children should receive 3 doses
using an adult formulation.
*Injecting drug users
Injecting drug users who have not been infected with hepatitis B should be
vaccinated.

158 The Australian Immunisation Handbook 9th Edition

Recipients of certain blood products


Screening of all blood donors for HBsAg has greatly decreased the incidence of
transfusion-related hepatitis B virus infection. However, patients with clotting
disorders who receive blood product concentrates have an elevated risk of
hepatitis B virus infection, and should therefore be vaccinated.
*Individuals with chronic liver disease and/or hepatitis C
Hepatitis B vaccination is recommended for those in this category who are
seronegative for hepatitis B.26
*Residents and staff of facilities for people with intellectual disabilities
Vaccination of carers, staff and susceptible residents is recommended in both
residential and non-residential care of people with intellectual disabilities.
Individuals adopting children from overseas

*Liver transplant recipients


If seronegative for hepatitis B, such individuals should be vaccinated
before transplantation as they may be at increased risk of infection from the
transplanted organ.
*Inmates and staff of long-term correctional facilities
Inmates are at risk of hepatitis B because of the prevalence of homosexual
intercourse, injecting drug use and amateur tattooing in some correctional
facilities. Therefore, they should be screened upon incarceration, and vaccinated
if susceptible.
Healthcare workers, ambulance personnel, dentists, embalmers, tattooists
and body-piercers
The risk to such workers differs considerably from setting to setting in different
parts of Australia, but it is recommended that all staff directly involved in patient
care,27 embalming, or in the handling of human blood or tissue, be vaccinated. In
addition, standard precautions against exposure to blood or body fluids should
be used as a matter of routine.
Others at risk
Police, members of the armed forces and emergency services staff should
be vaccinated if they are assigned to duties which may involve exposure.
Funeral workers and other workers who have regular contact with
human tissue, blood or body fluids and/or used needles or syringes.
People travelling to regions of intermediate or high endemicity, either
long-term or for frequent short terms, should be vaccinated.

Hepatitis B 159

3.6 Hepatitis B

These children should be tested for hepatitis B, and if they are HBsAg positive,
members of the adoptive family should be vaccinated.

Staff of child day-care centres will normally be at minimal risk of


hepatitisB. If advice on risk is sought, the enquiry should be directed to
the local public health authority.
Contact sports generally carry a low risk of hepatitis B infection.
Vaccination is nevertheless encouraged.
As the risk in Australian schools is very low,28 vaccination of classroom
contacts is seldom indicated. Nevertheless, vaccination of all children and
adolescents should be encouraged.
Sex industry workers.

(iv) Serological confirmation of post-vaccination immunity


Post-vaccination serological testing 4 to 8weeks after completion of the primary
course is recommended only for those in the following categories:
those at significant occupational risk (eg. healthcare workers whose work
involves frequent exposure to blood and body fluids),
those at risk of severe or complicated disease (eg. people with impaired
immunity, and individuals with pre-existing liver disease not related to
hepatitis B),
those in whom a poor response to hepatitis B vaccination is expected (eg.
haemodialysis patients),
sexual partners and household contacts of recently notified hepatitis B
carriers.29
Anti-HBs and HBsAg levels should be measured in infants born to known
HBsAg/HBeAg positive carrier mothers 3 to 12months after completing the
primary vaccine course. If anti-HBs levels are adequate and HBsAg is negative,
then children are considered to be protected.29

(v) Non-responders to primary vaccination


If adequate anti-HBs levels (10 mIU/mL) are not reached after the third dose,
the possibility of HBsAg carriage should be investigated. Those who are HBsAg
negative and do not respond should be offered further doses. These can be given
as either a fourth double dose or a further 3 doses atmonthly intervals, with
further testing at least 4weeks after the last dose.
There is limited evidence from several trials that HBsAg negative healthcare
workers, who are non-responders to a primary course of vaccination and
subsequent intramuscular booster schedule, as above, may respond to 5g of
Engerix-B (0.25mL of the adult formulation) administered intradermally at
fortnightly intervals (up to 4 doses) with anti-HBs levels measured before each
dose to assess for seroconversion.30-32 Persistent non-responders should be informed
that they are not protected and should minimise exposures, and about the need for
HBIG within 72 hours of parenteral exposure to HBV (see Table 3.6.3 Post-exposure
prophylaxis for non-immune individuals exposed to an HBsAg positive person).

160 The Australian Immunisation Handbook 9th Edition

Individuals who are at significant occupational risk who have a documented


history of a primary course of hepatitis B vaccine, but it is not known whether
they ever seroconverted, and they now have an antiHBs level <10 mIU/mL,
should be given a single booster dose of vaccine and have their anti-HBs level
checked 4weeks later. If the anti-HBs level is <10 mIU/mL, regard the individual
as a non-responder, give 2 further doses of hepatitis B vaccine atmonthly
intervals, and re-test for anti-HBs levels at least 4weeks after the last dose.

(vi) Booster doses

(vii) Interchangeability of vaccines


Although switching of brands is not recommended, in cases where the brand of
vaccine used for previous doses is not known, any age-appropriate formulation
may be used as there is no reason to believe that use of a different brand will
compromise immunogenicity or safety.

(viii) Post-exposure prophylaxis for hepatitis B


Following significant exposure (percutaneous, ocular, or mucous membrane) to
blood or potentially blood-contaminated secretions, the source individual should
be tested for HBsAg as soon as possible.
If the person exposed has not been previously vaccinated against hepatitis B, his/her
anti-HBs level and HBsAg should be determined immediately. If the person exposed
is anti-HBs negative, and the source is either HBsAg positive, or cannot be identified
and tested rapidly, administer a single dose of HBIG of 100 IU for children weighing
up to 30kg (about 5years of age) and 400 IU for all others, within 72 hours. Also
give hepatitis B vaccine (by IM injection into either the deltoid or anterolateral thigh,
depending on age) as soon as possible, but within 7days of exposure. Two further
doses of vaccine should be given, 1 and 6months after the first dose.
For previously vaccinated people exposed to either an HBsAg positive source
or a source whose hepatitis B status cannot be determined, post-exposure
prophylaxis is not necessary if there was a documented protective response (antiHBs level 10 mIU/mL) at any time after vaccination. If the response to previous
vaccination is unknown, the anti-HBs level should be determined as quickly as
possible. If the anti-HBs level is <10 IU/mL and HBsAg is negative, HBIG and
vaccine should be administered as above.

Hepatitis B 161

3.6 Hepatitis B

Although vaccine-induced antibody levels decline with time and may become
undetectable, booster doses are not recommended in immunocompetent
individuals after a primary course, as there is good evidence that a completed
primary course of hepatitis B vaccination provides long-lasting protection. This
applies to children and adults, including healthcare workers and dentists.33-39
However, booster doses are recommended for individuals with impaired
immunity, in particular those with either HIV infection or renal failure. The time
for boosting in such individuals should be decided by regular monitoring of antiHBs levels at 6 to 12-monthly intervals.33

In most instances, it is advisable to offer a course of hepatitis B vaccine to a nonimmune healthcare worker sustaining a needle-stick injury or other potential
hepatitis B exposure, since the injury or exposure itself is evidence that they work
in an area with a significant risk of exposure.
Table 3.6.3: Post-exposure prophylaxis for non-immune individuals exposed to
an HBsAg positive person
Type of exposure

Hepatitis B immunoglobulin

Vaccine

Perinatal
(exposure of
babies during
and after birth)

100 IU by IM
injection

Single dose
within 12
hours of birth,
preferably
immediately
after birth

0.5mL by IM
injection

Immediately
after birth
(preferably
within 24 hours,
no later than
7days*) then at
2, 4, and either
6 or 12months
of age

Percutaneous/
ocular
or mucous
membrane

400 IU by IM
injection

Single dose
within 72 hours

0.5mL or 1mL
by IM injection
depending
on age

Within 7days*
and at 1 and
6months after
first dose

Sexual

400 IU by IM
injection

100 IU if body
weight <30kg

Single dose
0.5mL or 1mL
within 14days
by IM injection
of sexual contact depending
on age

Within 14days*
and at 1 and
6months after
first dose

* The first dose can be given at the same time as HBIG, but should be administered at a
separate site.

Contraindications
The only absolute contraindications to hepatitis B vaccine are:
anaphylaxis following a previous dose of hepatitis B vaccine, or
anaphylaxis following any component of the vaccine.

Adverse events
Adverse events after hepatitis B vaccination are transient and minor, and
include soreness at the injection site (5%, common), fever (usually low
grade, 23%, common), nausea, dizziness, malaise, myalgia and arthralgia.
Fever can be expected in neonates immunised with hepatitis B vaccine
(0.63.7%, common).
Anaphylaxis has been reported very rarely in adults. Although various
adverse events such as demyelinating diseases, Guillain-Barr syndrome
and arthritis have been reported, there is no evidence of a causal relationship
with hepatitis B vaccination.40,41 There have been a few reports of generalised

162 The Australian Immunisation Handbook 9th Edition

febrile reactions attributed to yeast allergy, and exceptional instances of


polyarteritis nodosa have been reported.
The World Health Organization Global Advisory Committee on Vaccine
Safety states that multiple studies and review panels have concluded
that there is no link between MS [multiple sclerosis] and hepatitis B
vaccination.42
The vaccine produces neither therapeutic effects nor adverse events in
hepatitis B virus carriers. It is safe in those already immune to hepatitis B.

Use in pregnancy
Refer to Chapter 2.3, Groups with special vaccination requirements, Table 2.3.1
Vaccinations in pregnancy.

The product information for both Infanrix hexa and Infanrix Penta states that
these vaccines may be given as a booster dose at 18months of age. NHMRC
recommends that a booster dose of DTPa (or DTPa-containing vaccines) is not
necessary at 18month of age. However, DTPa-containing vaccine may be used
for catch-up of the primary schedule in children <8years of age.

Hepatitis B immunoglobulin (HBIG)


Hepatitis B immunoglobulin (HBIG) is prepared from plasma donated through
routine blood bank collection. Samples are selected on the basis that they contain
high levels of antibody to HBsAg. As stocks of HBIG are very limited, use should
be strictly reserved for those who are at high risk, such as babies born to hepatitis
B carrier mothers and healthcare workers who are exposed to the blood of
HbsAg positive individuals through occupational exposure. Requests should be
directed to the Australian Red Cross Blood Service in your State/Territory (see
Chapter 3.8, Immunoglobulin preparations Availability of immunoglobulins).
HBIG is given by IM injection.
Hepatitis B Immunoglobulin-VF CSL Bioplasma (160mg/mL
immunoglobulin (IgG) prepared from human plasma containing high levels
of antibody to surface antigen of the hepatitis B virus). 100 IU and 400 IU
ampoules, with the actual volume stated on the label on the vial.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

Hepatitis B 163

3.6 Hepatitis B

Variations from product information

3.7Human papillomavirus
Virology and HPV classification
Human papillomaviruses (HPVs) are small, non-enveloped viruses that have
circular double-stranded DNA. HPVs infect and replicate within cutaneous and
mucosal epithelial tissues, most commonly involving the skin or anogenital tract.
HPVs are designated as specific types according to sequence variation in the
major genes.
There are 40 distinct HPV genotypes that affect the genital tract; of these, 15
genotypes (16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 68, 73, 82) are designated
as high-risk as they are causally associated with the development of cervical
cancer. HPV genotypes 16 and 18 are the causative agents in 70 to 80% of
all cervical cancers. HPV genotypes 6 and 11 are among the HPV genotypes
designated as low-risk (for cancer), and are associated with 90% of genital warts
and 100% of recurrent respiratory papillomatosis (RRP) cases.1,2
High-risk genital HPV genotypes are associated with a spectrum of other
anogenital diseases, including vulval, vaginal, penile and anal cancers, and their
precursors. In addition, genital HPV genotypes are associated with extragenital
diseases, including some squamous cell carcinomas of the head and neck (highrisk HPV types) and recurrent respiratory papillomatosis (HPV types 6 and 11).
Persistent HPV infection is a necessary precursor of cervical cancer, but is not
sufficient in itself to cause the disease.3 For pre-cancerous lesions to form and
progress to cancer, the crucial event appears to be HPV DNA integration into the
host cell genome, which interferes with the expression and regulation of proteins
responsible for normal cell growth and repair.4 Malignancy due to build-up of
sufficient mutations for cellular transformation usually requires 10 to 20years,
but has been reported to occur in under 2years.5

Clinical features
HPV infection is often subclinical but, dependent upon the infecting HPV
genotype, may result in lesions that include cutaneous warts, genital warts,
cervical and other anogenital tract dysplasias and cancers, and respiratory
papillomatosis. Most genital HPV infections are cleared (no longer detectable)
within 12 to 24months (the median duration for high-risk genotypes is 7
to 10months).6-9 In a minority of infections, estimated at 3 to 10%, the virus
persists.10

164 The Australian Immunisation Handbook 9th Edition

Epidemiology
HPV infection
Transmission of HPV occurs through contact with infected skin or mucosal
surfaces, primarily via sexual contact for the genital HPV genotypes.
Transmission may rarely occur by other mechanisms, such as laryngeal infection
of infants during birth.11 There is a high probability of transmission following
sexual exposure to a person with a productive HPV infection, estimated to be
50 to 80%, after unprotected sexual intercourse.12-14 However, sexually active
adolescents and young adults may remain nave to all 4 vaccine HPV genotypes
or be infected with a non-vaccine HPV genotype.
HPV infection rates vary greatly between geographic regions, but it is estimated
that up to 79% of women worldwide will be infected with at least one genital
type of HPV at some point in their lives.15,16 HPV infection rates are highest
among young women, usually peaking soon after the age when most young
women become sexually active.17 Australian data show that, among women
currently aged 1619years, the median age of first intercourse is 16years.18

Human papillomavirus 165

3.7 Human papillomavirus

Although comprehensive risk-prediction models for HPV exposure are not


available, an increasing number of lifetime sex partners is consistently found
to be associated with HPV acquisition.19-24 A US population-based study of
women aged 1825years found genital HPV infection in 14.3% of women with
one lifetime sex partner, 22.3% with 2 lifetime sex partners, and 31.5% with
more than 3 lifetime partners.25 Australian women aged 1619years report a
median number of 2 lifetime sexual partners, women aged 2029years a median
of 4.3 lifetime sexual partners, and those aged 3039years a median of 4.7
lifetime sexual partners.26 Although its sensitivity is somewhat limited, HPV
seroprevalence measured using serum anti-HPV antibody levels can be used
to estimate cumulative lifetime exposure to specific types of HPV infection.27 In
a study of women in Finland, Dillner et al (1996) described a linear increase in
the risk of HPV16 seropositivity of 4% for every additional sex partner, ranging
from 4% for 1 lifetime partner to 35% among those with 6 or more partners.24
Similarly, HPV18 seroprevalence was observed to increase linearly at the rate of
3% per partner from 4% for 1 lifetime partner up to 24% for 6 or more partners.
In the US, population data indicate that 25% of women aged 2029years are
seropositive for HPV16.28 An increasing number of sexual contacts on the part of
their male partner is also associated with HPV acquisition in women.29

Cervical abnormalities
Cervical infection with HPV causes a range of pathological responses depending
on the genotype of HPV. These range from no reaction, carriage of HPV without
cytological changes, to a variety of cellular changes in the cervix. Histologically,
the cervical abnormalities have been referred to as cervical intraepithelial
neoplasia (CIN), with 3 grades of severity: CIN1 (mild dysplasia), CIN2
(moderate dysplasia) and CIN3 (severe dysplasia/carcinoma in situ). CIN3
and AIS (adenocarcinoma in situ) are immediate precursors of cervical cancer.
CIN2 represents a mix of low-grade and high-grade lesions (and hence it is
treated as a high-grade lesion). Cytologically, under the Australian Modified
Bethesda System, the term Low-grade Squamous Intraepithelial Lesion (LSIL)
encompasses changes thought to be due to HPV and mild dysplasia, and the
term High-grade Squamous Intraepithelial Lesion (HSIL) encompasses the
former categories of moderate dysplasia, severe dysplasia and carcinoma in
situ. Guidelines for the interpretation and treatment of screen-detected cervical
abnormalities are published by the NHMRC.12
Every year in Australia, approximately 90000 women have an LSIL detected
and 15000 women have an HSIL detected through Pap screening.12 The
incidence of both lesions peaks in women aged 2024years. In addition, there
are approximately 20000 hospital admissions per year for cervical dysplasia
and carcinoma in situ. This is an underestimate of the burden of disease, as
the investigation and management of cervical lesions are mostly carried out as
outpatient procedures, either in the private or public sector. For procedures in
the private sector, Medicare has processed, over the past 10years, claims for
an average of approximately 104000 examinations of the lower genital tract by
colposcopy and 14600 combined colposcopy procedures per year. As well as
physical side effects and complications from treatment for cervical abnormalities,
there is consistent evidence that receipt of an abnormal Pap smear result, and
the subsequent investigation and management, is associated with a considerable
psychosocial burden.30-35
It was originally thought that there was an inevitable progression from lowgrade abnormalities to high-grade abnormalities to cervical cancer. It is now
recognised that LSIL cytology is a manifestation of acute HPV infection, and
that most LSIL regresses over time.12 The absolute risk of cancer associated with
a high-grade abnormality is difficult to determine from available observational
data, but is estimated at less than 1% per year.36 Figure 3.7.1 summarises the
dynamic relationship between HPV infection and cervical health.

166 The Australian Immunisation Handbook 9th Edition

Figure 3.7.1: The dynamic relationship between HPV infection and cervical
health

Cervical cancer
Cancer of the uterine cervix is the second most common cause of cancer among
women worldwide.37 However, Australia has one of the lowest mortality rates
from cervical cancer in the world.38 In 2002, the age standardised incidence
rate in Australia was 6.8 per 100000 and, in 2004, the mortality rate was 1.9 per
100000, with an estimated 750 cases, 1800 hospitalisations and 250 deaths each
year from cervical cancer.39 This low incidence can be attributed to the success of
the National Cervical Screening Program, with cervical cancer in Australia now
occurring predominantly in unscreened or under-screened women. The largest
decline in cervical cancers has been observed for those of squamous origin,
with the incidence of adenocarcinomas being essentially unchanged. This has
been attributed to sampling difficulties in obtaining cells from the area where
adenocarcinoma arises, problems in pathological interpretation, and variations in
clinical investigation and treatment.12

Other anogenital cancers


High-risk HPV types (predominantly types 16 and 18) are also implicated in 50
to 90% of other anogenital cancers in both women and men, including cancers of
the vulva, vagina, anus, and penis, although these types have almost no role in

Human papillomavirus 167

3.7 Human papillomavirus

(Figure courtesy of the Australian Government Department of Health and Ageing.)

causing non-malignant lesions (see below). In Australia in 2001, there were 252
vulvar cancers (2.6 per 100000), 62 vaginal cancers (0.6 per 100000) and 225 anal
cancers (1.2 per 100000) diagnosed.40

Non-malignant lesions
Genital warts are a common manifestation of HPV type 6 and 11 infection.
Genital warts can cause significant psychological morbidity. In Australia, 4.0%
of men and 4.4% of women aged 1659years report ever being diagnosed
with genital warts.41 These prevalence estimates translate into approximately
36000 cases in Australia. The cumulative lifetime risk of genital warts has
been estimated at 10%.42,43 Peak attack rates occur in young women aged
1524years.44 An analysis of data from the BEACH cross-sectional survey
of national GP activity found that, between April 2000 and March 2003,
consultations for genital warts in women aged 1249years occurred at a rate of
0.17 per 100 encounters.45 Severe morbidity from genital warts, as measured by
hospitalisation, is uncommon and peaks in women 2024years of age (26 per
100000) (AIHW National Hospital Morbidity Database 2006). Morbidity not
causing hospitalisation includes recurrence and a range of local complications.
Worldwide, the best epidemiological data on genital wart incidence comes from
the United Kingdom.2
Exposure to HPV types 6 and 11 at birth can also cause recurrent respiratory
papillomatosis in children. This relatively rare disease, with an estimated
incidence of 4 per 100000 children,46,47 is characterised by repeated growth of
warts in the respiratory tract requiring repeated surgery. Adults can also develop
recurrent respiratory papillomatosis.

Type-specific HPV epidemiology


Worldwide, approximately 50% of cervical cancers contain HPV16 DNA and 16%
contain HPV18 DNA.48,49 Among cervical adenocarcinomas, 70% contain HPV
16 or 18 DNA, with HPV18 being relatively more common (37.7%) than HPV16
(31.3%).48 HPV16 has been detected in 48% of HSILs, 19% of LSILs and 2% of
cytologically normal women. HPV18 has been detected in 7% of HSILs, 6% of
LSILs and 0.7% of cytologically normal women.50-52 The low-risk HPV types 6
and 11 have been detected in 6.2% and 3.2% of LSILs respectively and each type
in 0.1% of cytologically normal women.51,52
Australian studies indicate that the 5 most frequent HPV genotypes identified in
553 cervical cancers were HPV16 (60%), HPV18 (20%), HPV45 (5%), and HPV39
and HPV73 (2.3% each).53-57 Best available Australian data indicate that HPV16
and HPV18 are, respectively, responsible for approximately 60%/20% of cervical
cancers and 37%/8% of high-grade cervical abnormalities.53,54

168 The Australian Immunisation Handbook 9th Edition

Vaccines
HPV vaccines have been developed using recombinant DNA technology based
on virus-like particles (VLPs), which are not infectious and do not have any
cancer-causing potential.
There are 2 HPV vaccines registered for use in Australia. The bivalent vaccine,
2vHPV vaccine (CERVARIX), contains VLPs of HPV genotypes 16 and 18, and
is administered as a 3-dose schedule at 0, 1 and 6months. The quadrivalent
vaccine, 4vHPV vaccine (GARDASIL), is administered at 0, 2 and 6months and
contains VLPs of HPV genotypes 16, 18, 6 and 11.
CERVARIX GlaxoSmithKline (human papillomavirus vaccine
recombinant protein particulate (VLP) vaccine containing the major capsid
(L1) protein of HPV types 16 and 18). Each 0.5mL monodose pre-filled
syringe or vial contains 20g each of HPV types 16 and 18 adjuvanted
with AS04 (AS04 is comprised of 500g aluminium hydroxide and 50g of
3-O-desacyl-4-monophosphoryl lipid A [MPL]); 4.4mg sodium chloride;
624g sodium dihydrogen phosphate dihydrate.

It is important to note that HPV vaccines are prophylactic vaccines (ie. designed
to prevent initial HPV infection). In women who are already infected with HPV
types covered by the vaccines before vaccination (ie. HPV DNA positive), the
vaccines do not treat infection or prevent disease caused by that type.58
In women HPV DNA negative and HPV seronegative for relevant types, both
vaccines are highly effective at preventing persistent type-specific infection and
related cervical disease (~90100%).59-62 The 4vHPV vaccine also has established
efficacy against external genital lesions (warts, and vulval and vaginal
dysplasias) in women. Vaccine efficacy against external genital lesions related to
HPV 6, 11, 16 or 18 in women who were nave to vaccine types at the beginning
of the trials and who received 3 doses was 99% (95% CI: 95100%).
Compared to HPV DNA negative and HPV seronegative women, vaccine efficacy
(VE) in women who received vaccine regardless of HPV status at baseline
and may, therefore, have had previous infection, was much lower. Against
HPV16/18-related CIN2/3 or worse, VE was 44% (95% CI: 3155%) at a mean
of 3years follow-up63 and against high-grade CIN caused by any HPV type it

Human papillomavirus 169

3.7 Human papillomavirus

GARDASIL CSL Biotherapies/Merck & Co Inc (human papillomavirus


vaccine recombinant protein particulate (VLP) vaccine containing the
major capsid (L1) protein of HPV types 6, 11, 16 and 18). Each 0.5mL
monodose pre-filled syringe or vial contains 20, 40, 40 and 20g of HPV
types 6, 11, 16 and 18, respectively, adsorbed onto 225g aluminium
hydroxyphosphate sulphate; 9.65mg of sodium chloride; 780g of
L-histidine; 50g of polysorbate 80; 35g of sodium borate. May also
contain yeast proteins.

was 18% (95% CI: 729%).63 However, vaccine efficacy is expected to be higher
over a longer duration of follow-up, as the proportion of disease due to incident
infection (where vaccination has an effect) increases compared to the proportion
of disease due to infection/disease at baseline (which is not affected by the
vaccine). These data reflect the reduced impact of vaccinating women in whom a
proportion will already have been infected with HPV, eg. older women who are/
have been sexually active. Vaccine efficacy estimates in populations including
women already infected with HPV at baseline have not yet been published for
2vHPV vaccine, but a similarly reduced impact, compared with an HPV nave
population, can be anticipated.
It is possible that HPV vaccines may provide some protective efficacy against
disease due to types closely related to types 16 and 18, in particular HPV31 and
HPV45, but published data supporting this hypothesis are currently limited to
infection endpoints only and are imprecise.60,64-66
When given as a 3-dose series, HPV vaccines elicit neutralising antibody titres
many times higher than those observed following natural infection.67-69 Antibody
responses peak atmonth 7 (1month after dose 3) at titres between 7 and 150
times greater than following natural infection, depending upon the HPV type
and vaccine.59,62,70,71 Following an initial decline, they appear to plateau at 18
to 24months, remaining stable for at least 5years at levels above or at least
equivalent to those seen following natural infection.58,60,62,70 It should be noted
that there is no standard serological assay for detecting HPV antibodies and
no protective titre has been established. Therefore, absolute titres achieved (as
reported in the randomised trials) are not directly comparable between 2vHPV
and 4vHPV vaccines. Similarly, differences in methodologies and the populations
examined make direct comparisons of published 4vHPV and 2vHPV vaccine
efficacy estimates difficult.
Overall, seroconversion occurs in 99 to 100% of those vaccinated.58,60,61 The duration
of immunity after vaccination is not yet known (but is of at least 5years duration);
hence, it is possible that booster doses may be required in the future.58,60,62
There are currently no clinical efficacy data available in males or in pre-adolescent
females (as collection of genital specimens is not appropriate). Antibody response
to both 4vHPV and 2vHPV vaccination has been evaluated in pre-adolescent
and adolescent females (915years of age and 1014years of age, respectively).
In males, antibody response has only been studied for 4vHPV, and only in
the age group 915years, through immunological bridging studies.72,73 Young
males and females administered 4vHPV vaccine and females aged 1014years
administered 2vHPV vaccine produce antibody responses that are at least 2-fold
higher compared to women in whom clinical efficacy has been demonstrated.
The peak antibody levels achieved following vaccination decrease with age.
Immunobridging studies for older women (aged >26 45years) administered
the 2vHPV demonstrate antibody titres in a comparable range to women in the
1525 year age group who are in the plateau phase of long-term follow-up.

170 The Australian Immunisation Handbook 9th Edition

These data were current at the time of publishing The Australian Immunisation
Handbook.

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.74 Store at
+2C to +8C. Do not freeze. Protect from light.

Dosage and administration


The dose of 2vHPV vaccine is 0.5mL administered by IM injection. The
recommended schedule is 0, 1 and 6months. The second dose of 2vHPV can be
administered between 1 and 2.5months after the first dose.
The dose of 4vHPV vaccine is 0.5mL administered by IM injection. The
recommended schedule is 0, 2 and 6months. In clinical studies, efficacy for
4vHPV vaccine has been demonstrated in individuals who have received all
3 doses within a 1 year period. Where flexibility in the recommended dosing
schedule is unavoidable, the second dose should be administered at least
1month after the first dose and the third dose should be administered at least
3months after the second. There is no need to repeat earlier doses. Give missing
dose(s) as soon as is practicable, making efforts to complete doses within
12months.

There are no clinical data regarding concomitant administration of either


2vHPV or 4vHPV vaccine with adolescent/adult formulation dTpa or varicella
vaccine, but there is no reason to expect any adverse outcomes if they are given
simultaneously, using different injection sites.

Recommendations
Both vaccines are recommended to provide protection against oncogenic HPV
type 16 and/or 18 cervical disease. If protection against genital warts is desired,
the 4vHPV vaccine provides protection against HPV types 6 and 11, which are
associated with more than 90% of these lesions.67 (See also Vaccines above.)

(i) Females aged 1013years (Safety-Grade B)(Efficacyno data)(Immunogenicity-Grade B)67


HPV vaccine is recommended for females 1013years of age. Currently only the
4vHPV vaccine is on the NIP schedule for females aged 1213years. Please refer
to your State/Territory health authority for further information (see Appendix
1, Contact details for Australian, State and Territory Government health authorities and
communicable disease control).

Human papillomavirus 171

3.7 Human papillomavirus

4vHPV vaccine has been administered concomitantly with hepatitis B vaccine in


clinical trials, with no reduction in immunogenicity of either vaccine observed.

(ii) Females aged 1418years (Safety-Grade B)(EfficacyGrade B)(Immunogenicity-Grade B)67


HPV vaccine is also recommended for females 1418years of age. While some
females in this age group will already have commenced sexual activity, the
majority will not yet be infected with a HPV vaccine type.

(iii) Females aged 1926years (Safety-Grade A)(EfficacyGrade A)(Immunogenicity-Grade A)67


HPV vaccine is also recommended for females 1926years of age.
In females in this age group who have never had sexual intercourse the vaccine
efficacy will be comparable to younger women, and HPV vaccination is
recommended. In sexually active females 1926years of age, the overall benefit
from HPV vaccination is likely to be less; however, past or current infection with
all HPV types covered by the vaccine is unlikely.
NB. The absolute benefit of HPV vaccine to an individual sexually active woman
cannot be determined clinically, as appropriate tests to detect both previous and
current HPV infection with vaccine types are not available.
In all sexually active women, the most important preventive intervention against
cervical disease remains regular Pap screening. Vaccination is not an alternative
to Pap screening but is complementary. The National Cervical Screening Program
recommends routine screening with Pap smears every 2years for all women
between the ages of 18 (or 2years after first sexual intercourse, if later) and
69years.

(iv) Females aged 27years (Safety 2vHPV vaccine-Grade B; 4vHPV vaccine-no


data)(Efficacy-no data)(Immunogenicity 2vHPV-Grade B; 4vHPV-no data)67
2vHPV vaccine is registered for use in females 2745years of age on the basis
of safety and bridging immunogenicity data. The extent of benefit that can
be expected to be derived from the use of HPV vaccine in this age group will
depend upon past sexual history and the likelihood of new sexual partners in the
future (ie. an assessment of likely past and future HPV exposure) and the sexual
behaviour of her male partner(s). HPV-related cervical infection and Pap test
abnormalities peak in women aged <30years in Australia.
4vHPV vaccine is not registered for use in females over the age of 27years as
there are no safety or efficacy data to support its use in this age group.
In all sexually active women, the most important preventive intervention against
cervical disease remains regular Pap screening. Vaccination is not an alternative
to Pap screening but is complementary. The National Cervical Screening Program
recommends routine screening with Pap smears every 2years for all women
between the ages of 18 (or 2years after first sexual intercourse, if later) and
69years.

172 The Australian Immunisation Handbook 9th Edition

For women who have recently been diagnosed with cervical dysplasia, or have
been treated for this in the past, HPV vaccine will have no impact on current
disease, but may prevent future dysplasia due to a different HPV vaccine type.

(v) Males [For males aged 915years (Safety 4 vHPV vaccineGrade B; 2vHPV vaccine-no data)(Efficacy-no data)(Immunogenicity
4vHPV vaccine-Grade B; 2vHPV vaccine-no data)]67
4vHPV vaccine is licensed for use in males aged 915years. 4vHPV vaccine
produces high antibody titres in pre-adolescent and adolescent males but it is
not known whether vaccination of males can either prevent transmission of HPV
or provide protection against genital HPV infection, genital warts, anogenital
dysplasia or anogenital cancers. 2vHPV vaccine is not registered for use in males.
There is no recommendation for vaccination of males at this time due to the lack
of clinical efficacy data.

Contraindications
The only absolute contraindications to HPV vaccine are:
anaphylaxis following a previous dose of the vaccine, or
anaphylaxis to any vaccine component. The 4vHPV vaccine may contain
minute amounts of yeast proteins.

Precautions
There are limited clinical trial data available for this group. However, as HPV
vaccines are not live vaccines, they can be administered to women who are
immunosuppressed as a result of disease or medications. The immune response
and vaccine efficacy might be less than in individuals who are immunocompetent
(see Chapter 2.3, Subsection 2.3.3, Vaccination of individuals with impaired immunity
due to disease or treatment).

Adverse events
Both the 2vHPV and 4vHPV vaccines are generally safe and well tolerated. A
variety of comparators were used in clinical trials of 2vHPV and 4vHPV, but
data comparing vaccine adverse events with an aluminium-containing placebo
are available for both vaccines and are quoted below for common local adverse
reactions. More detailed information about adverse events occurring in the
vaccine trials is available from the product information for 2vHPV vaccine and
from the US FDA for 4vHPV.75
In clinical trials of the 2vHPV vaccine, the most commonly reported adverse
events were injection site pain 78%, swelling ~26% and erythema ~30%
compared to ~53%, ~8% and 11% in the aluminium hydroxide placebo group.
Incidence of injection site pain decreased across the 3 doses, whereas there was

Human papillomavirus 173

3.7 Human papillomavirus

People with impaired immunity

a slight increase in the reported proportion with swelling and erythema after
successive doses.
In clinical trials of the 4vHPV vaccine the most commonly reported adverse
events were injection site pain ~81%, swelling ~24% and erythema ~24%,
compared to ~75%, ~16% and ~18% in the aluminium-containing placebo group.
The incidence of injection site pain was approximately equal across the 3 doses,
whereas there was a modest increase in the reported proportion with swelling
and erythema after successive doses.
HPV vaccines are well tolerated by those who have already been exposed to the
HPV types included in the vaccine.

Use in pregnancy
HPV vaccine should not be given during pregnancy (see Chapter 2.3, Subsection
2.3.2, Vaccination of women planning pregnancy, pregnant or breastfeeding women, and
preterm infants).
It should be noted that there is no evidence from animal studies, or among HPV
vaccine trial participants who inadvertently became pregnant, of teratogenicity
or of adverse fetal outcomes and, therefore, HPV vaccination during pregnancy
is not an indication for termination.
Where vaccine has inadvertently been administered during pregnancy, further
doses should be deferred until after delivery.

Use during lactation


HPV vaccine may be given while lactating (see Chapter 2.3, Subsection 2.3.2,
Vaccination of women planning pregnancy, pregnant or breastfeeding women, and
preterm infants).
In trials, 995 nursing mothers received 4vHPV vaccine or placebo, and no relation
between vaccination and adverse events was observed. The effect on breastfed
infants of the administration of 2vHPV vaccine to their mothers has not been
evaluated in clinical studies. It is not known whether HPV vaccine antigens or
HPV antibodies are excreted in human milk.

Variations from product information


None.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

174 The Australian Immunisation Handbook 9th Edition

3.8 Immunoglobulin preparations


Introduction
Passive immunity can be provided by administration of human
immunoglobulin.1-3 The protection afforded is immediate, but is transient
and lasts for only a fewweeks, as the half-life of IgG, the major constituent, is
between 3 and 4weeks.
There are 2 types of immunoglobulin, normal and specific. It is important
to recognise that separate immunoglobulin preparations are provided for
intramuscular (IM) use and for intravenous (IV) use. These have different
properties, and the preparations should be given only by the recommended
route. Administration of IM immunoglobulin by the IV route will lead to
severe reactions.

Normal human immunoglobulin (NHIG)


This is derived from the pooled plasma of blood donors. It contains antibody to
microbial agents which are prevalent in the general population.

Specific immunoglobulins
Specific immunoglobulin preparations are obtained from pooled blood
donations from patients convalescing from the relevant infection, donors recently
vaccinated with the relevant vaccine, or those who, on screening, have been
found to have sufficiently high antibody concentrations. These blood-derived
specific immunoglobulins therefore contain concentrations of antibody to an
individual organism or toxin at a higher titre than would be present in normal
immunoglobulin.

Potential interaction with vaccines


Live attenuated virus vaccines
Immunoglobulin preparations can interfere with the response to live
attenuated virus vaccines by preventing vaccine strain viral replication after
vaccine administration. Therefore, administration of live attenuated virus
vaccines, such as measles and varicella vaccines, should be deferred for at

Immunoglobulin preparations 175

3.8 Immunoglobulin
preparations

Donors of blood used for the production of NHIG and specific immunoglobulin
products are screened and products treated to minimise the risk of the
immunoglobulin preparations containing HIV, hepatitis A, hepatitis B or
hepatitis C viruses, or parvovirus. Two dedicated pathogen inactivation steps
are incorporated into the manufacturing process. A pasteurisation step is usually
used during manufacture. The risk of prion transmission remains theoretical.

least 3months after the IM administration of NHIG, and for at least 9months
after the administration of NHIG (intravenous).4 For the same reason,
administration of immunoglobulin products should be deferred if possible
until at least 2weeks after a vaccine has been given, unless it is essential that
immunoglobulin be given. However, Rh (D) immunoglobulin (anti-D) does
not interfere with the antibody response to MMR vaccines and the two may
be given at the same time in different sites with separate syringes or at any
time in relation to each other (see Chapter 2.3, Groups with special vaccination
requirements, Table 2.3.5 Recommended intervals between either immunoglobulins
or blood products and MMR, MMRV or varicella vaccination).

Inactivated vaccines
Inactivated vaccines such as tetanus, hepatitis B or rabies may be
administered concurrently with immunoglobulin preparations, or at any time
after, using separate syringes and separate injection sites to induce passive/
active immunity. This usually would occur when there has been actual or
possible acute exposure.

Availability of immunoglobulins
CSL Bioplasma supplies NHIG for IM use. Rabies immunoglobulin can be
obtained only upon application from State/Territory health authorities.
Respiratory syncytial virus (RSV) monoclonal antibody (Synagis; Abbott
Australia) is available commercially.
The specific immunoglobulins and the CSL Bioplasma NHIG for IV use, which
are derived from Australian donated plasma, can be obtained only from the
Australian Red Cross Blood Service (ARCBS) with permission from a ARCBS
medical officer. The Red Cross Blood Service can be contacted by telephone (ACT
02 6206 6006; NSW 02 9229 4444; NT 08 8927 7855; QLD 07 3835 1333; SA 08 8422
1200; TAS 03 6230 6230; VIC 03 9694 0111; WA 08 9325 3333). The Australian Red
Cross Blood Service supplies these products free of charge.

Transport, storage and handling


All immunoglobulins must be protected from light and stored at +2C to +8C.
Do not freeze.

Normal human immunoglobulin (NHIG) intramuscular use


NHIG is prepared by plasma fractionation of blood collected from volunteer
donors by the Australian Red Cross Blood Service. It is a sterile solution of
immunoglobulin, mainly IgG, and contains those antibodies commonly present
in adult human blood. In Australia, NHIG is supplied as a 16% solution, in the
United States as a 16.5% solution, and in the United Kingdom as a 10% solution.

176 The Australian Immunisation Handbook 9th Edition

Normal Immunoglobulin-VF (human) (NHIG) CSL Bioplasma. A


sterile preservative-free solution of immunoglobulin G (IgG) 160mg/mL
prepared from Australian blood donations and made available through the
Australian Red Cross Blood Service. It is supplied in 2mL and 5mL vials
for IM injection.

Administration
NHIG should be given by deep IM injection using a large (19 or 20) gauge
needle. The NHIG should be introduced slowly into the muscle, to reduce pain.
This product should not be administered intravenously because of possible
severe adverse events, and hence an attempt to draw back on the syringe after
IM insertion of the needle should be made in order to ensure that the needle is
not in a small vessel. A special product for IV use (NHIG (intravenous)) has been
developed for patients requiring large doses of immunoglobulin.

Recommendations
Immunoglobulin preparations may be given to susceptible individuals as either
pre-exposure or post-exposure prophylaxis against specific infections. Normal
pooled immunoglobulin contains sufficiently high antibody concentrations to
be effective against hepatitis A and measles. The duration of effect of NHIG is
dose-related. It is estimated that protection is maintained for 3 to 4weeks with
standard recommended doses of NHIG.

(i) Prevention of hepatitis A (see also Chapter 3.5, Hepatitis A)


NHIG contains sufficiently high levels of antibody against hepatitis A to be able
to prevent or ameliorate infection in susceptible individuals,5 if administered
within 2weeks of exposure.6
Because the hepatitis A vaccine is readily available, there is no place for the
routine use of NHIG to prevent hepatitis A in travellers. It should be given
(at the same time as a dose of hepatitis A vaccine) only to those, such as nonimmune aid-workers to be deployed within 2weeks, who will be living in very
inadequate circumstances.7,8

(ii) Prevention of hepatitis B

(iii) Prevention of measles (see also Chapter 3.11, Measles)


NHIG contains a sufficiently high concentration of antibody against measles
to be able to prevent or ameliorate infection in susceptible individuals. NHIG
should be given as soon as possible and within 7days of exposure. Passive
protection against measles particularly may be required if the exposed individual
has an underlying immunological disorder (HIV/AIDS, immunosuppressive

Immunoglobulin preparations 177

3.8 Immunoglobulin
preparations

See Chapter 3.6, Hepatitis B, under Management of infants born to hepatitis B


carrier mothers and Post-exposure prophylaxis for hepatitis B.

therapy), or to control an outbreak of measles among non-immunised


individuals, eg. in a childcare centre. The use of NHIG should be considered in
HIV-positive individuals exposed to a patient with measles.

(iv) Prevention of varicella (see also Chapter 3.24, Varicella)


Zoster immunoglobulin (ZIG) is able to prevent or ameliorate varicella in infants
<1month of age, in children who are being treated with immunosuppressive
therapy, and in pregnant women.9,10 ZIG should be given as soon as possible, and
preferably within 96 hours, after exposure. ZIG is recommended for non-immune
HIV-positive individuals up to 7days after exposure to clinical cases of either
varicella or zoster.
If ZIG is unavailable, large doses of NHIG can be given intramuscularly. This
does not necessarily prevent varicella, but it lessens the severity of the disease.
The dose of NHIG is 0.41.0mL perkg body weight given by the IM route.

(v) Immune deficiency


Patients with abnormal antibody production (primary
hypogammaglobulinaemia, multiple myeloma, chronic lymphoblastic
leukaemia) are usually treated with the IV preparation of normal human
immunoglobulin (NHIG (intravenous)).2
However, in some cases, NHIG is given by IM injection in a dose of 400600mg/
kg (0.40.6 g/kg) every 2 to 4weeks. The aim of therapy is to maintain serum IgG
levels above 6 g/L. Some patients may receive the IM (160mg/mL) preparation
subcutaneously.
NB. Skin tests with NHIG should not be undertaken. The intradermal injection of
concentrated immunoglobulin causes a localised area of inflammation which can
be misinterpreted as a positive allergic reaction. True allergic responses to NHIG
given by IM injection are extremely rare.

Contraindications
Hypersensitivity reactions occur rarely but may be more common in patients
receiving repeated injections. It is recommended that NHIG should not be
given to individuals with absolute IgA deficiency, as the small amounts of IgA
in NHIG could theoretically lead to the development of anti-IgA antibodies in
these individuals. NHIG should not be administered to individuals who have
severe thrombocytopenia or any coagulation disorder that would contraindicate
intramuscular injections.

Adverse events and precautions


Local tenderness, erythema and muscle stiffness at the site of injection sometimes
occurs and may persist for several hours after injection. Systemic adverse events
such as mild pyrexia, malaise, drowsiness, urticaria and angioedema may occur
occasionally. Skin lesions, headache, dizziness, nausea, general hypersensitivity
reactions and convulsions may occur rarely.

178 The Australian Immunisation Handbook 9th Edition

Anaphylaxis following an injection of NHIG is very rare, but has been reported.
Anaphylaxis is more likely to occur if NHIG for IM use is inadvertently given
intravenously.

Normal human immunoglobulin (NHIG) intravenous use


Normal human immunoglobulin (intravenous) is usually abbreviated as NHIG
(intravenous).
Intragam P CSL Bioplasma. A sterile preservative-free solution of
immunoglobulin G (IgG) 60mg/mL prepared from Australian blood
donations and made available through the Australian Red Cross Blood
Service. Intragam P contains only trace amounts of IgA, and the final
solution contains 100mg/mL maltose. It is supplied as 3 g in 50mL and 12
g in 200mL bottles (for intravenous use).
Sandoglobulin NF liquid CSL Bioplasma (sterile preservative-free
solution of immunoglobulin G (IgG), 120mg/mL). It is supplied as 6 g in
50mL and 12 g in 100mL bottles (for intravenous use). A sterile lyophilised
preparation for reconstitution, containing human gammaglobulin. The
product is reconstituted with sodium chloride 0.9% solution to a sterile 3%
or 6% solution. Available in 6 g vials (for intravenous use).
Octagam Octapharma. A sterile preservative-free solution of
immunoglobulin G (IgG) 50mg/mL prepared from multiple blood donors.
It is supplied as 1 g in 20mL vials, and as bottles of 2.5 g in 50mL, 5 g in
100mL and 10 g in 200mL (for intravenous use).
The available NHIG (intravenous) preparations in Australia have different
recommendations for dosage and administration and the product information
must be consulted before the use of each individual product. The text below
provides an overview of dosage and administration for NHIG (intravenous).

Dosage and administration

The dose for replacement therapy in individuals with immune deficiency is


0.40.6 g/kg every 3 to 4weeks. In Kawasaki disease, a single dose of 2 g/kg
given over at least 6 to 8 hours is recommended, repeated once if fever fails to
resolve within 48 hours. Doses should be calculated to the nearest (next highest)
bottle so as not to waste any immunoglobulin. Giving slightly more than the
calculated dose per kilogram will not be harmful.

Immunoglobulin preparations 179

3.8 Immunoglobulin
preparations

The infusion should be commenced slowly and the rate increased gradually.
Patients should be closely observed for the duration of the infusion. The patients
pulse, blood pressure and respiration rate should be recorded at 15-minute
intervals, and their temperature every hour. All these observations should also be
made and recorded before the commencement of the infusion.

Recommendations
(i) Antibody deficiency disorders
NHIG (intravenous) is indicated for patients with antibody deficiency disorders
requiring large doses of immunoglobulin. Therapy in these patients is usually
administered atmonthly intervals. NHIG (intravenous) produces higher serum
concentrations of IgG after administration than the IM preparation.2

(ii) Kawasaki disease


In clinical studies, NHIG (intravenous) has been found to be effective in the acute
phase of Kawasaki disease, as it reduces the risk of coronary artery involvement,
and is associated with more rapid resolution of other acute phase features of the
disease.11-15

(iii) Other uses


NHIG (intravenous) has been used in the management of immune
thrombocytopenia,16 Guillain-Barr syndrome,17,18 chronic inflammatory
demyelinating polyneuropathy,18 toxic shock syndrome,19 post-transfusion
purpura, in patients with bacterial infections associated with secondary
immunodeficiency, and in other inflammatory and infective disorders.3
(NB. Some recommendations in this section are not included in the current
registered indications for Intragam P. Potential users of NHIG (intravenous) in
these circumstances should consult the Australian Red Cross Blood Service or the
manufacturer.)

Contraindications
Individuals who are known to have had an anaphylactic or severe systemic
response to NHIG should not receive further immunoglobulin. Individuals with
selective IgA deficiency should not receive immunoglobulin preparations.

Adverse events and precautions


Adverse events with NHIG (intravenous) consist of shivering, headache, chest
and back pains and moderate pyrexia. Severe headache, sometimes attributed
to aseptic meningitis, has also been observed with NHIG (intravenous). This
can be ameliorated by slowing the infusion or by mixing the 60mg/mL
preparation with an equal volume of normal saline before administration.
There have been isolated reports of renal dysfunction and acute renal failure
following the administration of NHIG (intravenous). To date, anaphylactic
shock has not been experienced with NHIG (intravenous). Subjects with
absolute selective IgA deficiency have an increased risk of severe adverse
events following NHIG (intravenous).

180 The Australian Immunisation Handbook 9th Edition

Specific immunoglobulins
These products are used to protect individuals against specific microbial agents
such as hepatitis B,20 rabies and varicella-zoster viruses,9,10 and tetanus toxin.
Each of these specific immunoglobulins is described in more detail in this
Handbook in the chapter or section relevant to these specific infections.
In addition, specific immunoglobulins are available for botulism,
cytomegalovirus (CMV) and respiratory syncytial virus (RSV) as described
below. Adverse events and storage requirements for these specific
immunoglobulins are similar to those for NHIG (IM) and, therefore, are not
repeated here.

Botulism antitoxin (formerly known as


Botulism Immune Globulin, BIG)
Equine antitoxin made in horses has long been used in the treatment of adult
botulism, but has not been shown to be effective in infant botulism.21
Equine antitoxin is manufactured by major vaccine producing companies such as
Chiron. Use in Australia is governed by the Therapeutic Goods Administrations
Special Access Scheme and physicians wishing to access this stock should
initially contact their State/Territory health authority. Hypersensitivity,
presenting as fever, serum sickness or anaphylaxis, may follow its use. Skin
testing followed by appropriate dosing should be administered according to the
manufacturers instructions.
A new intravenous botulinum antitoxin, produced in the USA, reduced the
duration of mechanical ventilation and hospitalisation significantly in infant
botulism.22 It is not currently registered in Australia, but is registered by the US
FDA. The sponsor is Californian Department of Health Services. Access to this
product should be sought through the Special Access Scheme.

CMV immunoglobulin

The product contains no antibacterial agent, and so it must be used immediately


after opening. Any unused portion must be discarded. If the solution has been
frozen, it must not be used. If the use of CMV immunoglobulin is contemplated,
detailed protocols for administration and management of adverse events should
be consulted, in addition to the Product Information.

Immunoglobulin preparations 181

3.8 Immunoglobulin
preparations

CMV immunoglobulin is indicated for the prevention of CMV infection in


immunodeficient people at high risk of severe CMV infection, such as after bone
marrow and renal transplants.23-31 The treatment of established CMV infection is
primarily with antivirals such as ganciclovir, and there is scant evidence that the
addition of CMV immunoglobulin improves outcome.25,26,28 It would seem most
logical to reserve the use of CMV immunoglobulin to treat established CMV
infection in those patients with hypogammaglobulinaemia.

CMV Immunoglobulin-VF (human) CSL Bioplasma (sterile solution of


immunoglobulin prepared from human plasma containing high levels of
antibody to CMV). The plasma protein content is approximately 60mg/mL
of which at least 98% is IgG immunoglobulin with a CMV immunoglobulin
activity of 1.5 million CMV units per vial. Maltose is added to achieve
isotonicity.

RSV immunoglobulin
Several clinical studies of immunoglobulin against RSV have been conducted
overseas using hyperimmune polyclonal RSV immunoglobulin (RSVIG) derived
from blood donations.32-34 It has been shown to reduce the incidence and severity
of RSV infections when given prophylactically in some babies and infants at
high risk of severe infection. Benefit has been shown for babies and infants with
bronchopulmonary dysplasia (BPD), for those with prematurity without BPD,
and children with haemodynamically significant congenital heart disease.34,35
RSVIG has caused severe cyanotic episodes and poor outcome after surgery in
children with congenital heart disease and is contraindicated in such children.36
RSVIG is not registered in Australia.
A humanised mouse monoclonal antibody to RSV produced by cultured cells
palivizumab is now registered in Australia for prevention of serious lower
respiratory tract disease caused by RSV in children at high risk of RSV disease.
This product is given by IM injection eachmonth during periods of anticipated
risk of RSV. Palivizumab was found to reduce the absolute risk of hospitalisation
from about 10% to about 5% for babies born prematurely,37 for babies with
BPD,37 and also for babies with haemodynamically significant congenital heart
disease.35 It has not been shown to reduce the incidence of more severe outcomes
such as the need for ventilation, nor has it been shown to reduce mortality.35,37
Palivizumab is more effective and less costly than RSVIG, but its cost is still
prohibitive. Cost-effectiveness analyses have not shown palivizumab to be costbeneficial, and even analysis of sub-groups of children at high risk has not shown
a single subgroup where prophylaxis results in net savings.38,39
Synagis Abbott Australia (palivizumab). Supplied in single-use vials of
powder, to be reconstituted with sterile water for injection; 50mg in 4mL
vial; 100mg in 10mL vial.

182 The Australian Immunisation Handbook 9th Edition

Dosage and administration


Palivizumab is administered by IM injection preferably in the anterolateral thigh,
in a dose of 15mg/kg once amonth. Where possible, the first dose should be
administered before commencement of the RSV season.

Use in pregnancy
Refer to Chapter 2.3, Groups with special vaccination requirements, Table 2.3.1
Vaccinations in pregnancy.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

3.8 Immunoglobulin
preparations

Immunoglobulin preparations 183

3.9 Influenza
Virology
The influenza viruses are orthomyxoviruses. They are classified antigenically
as types A, B or C, but only influenza A and B are clinically important in
human disease.1 Influenza viruses possess 2 surface glycoprotein antigens,
the haemagglutinin (H) which is involved in cell attachment during infection,
and the neuraminidase (N) which facilitates the release of newly synthesised
virus from the cell. The influenza A viruses can be segregated into subtypes
based on differences in these surface antigens, whereas influenza B cannot be
segregated into subtypes. Antibody against the surface antigens, particularly the
haemagglutinin, reduces infection or severe illness due to influenza.
Both influenza A and influenza B viruses undergo frequent changes in their
surface antigens. Both influenza A and B undergo stepwise mutations of genes
coding for H and N. This results in cumulative changes in influenza antigens, or
antigenic drift. This is responsible for the annual outbreaks and epidemics of
influenza and is the reason that the composition of influenza vaccines requires
annual review. Antigenic shift, defined as a dramatic change in H antigen with
or without a similar change in N, occurs occasionally and unpredictably and
can cause pandemic influenza.1 Pandemic subtypes arise spontaneously from
antigenic shift or as a result of genetic reassortment (mixing) between bird
(avian) or animal viruses and human strains.
Three pandemics are recognised in the 20th century, in 1918 (H1N1), 1957
(H2N2) and 1968 (H3N2). These pandemic strains have gone on to circulate in
the community, with various subtypes causing seasonal influenza and, since
1977, 2 subtypes of influenza A, A (H1N1) and A (H3N2), co-circulating in the
human population together with influenza B. Recently, the avian influenza
virus subtypes, A (H5N1) and A (H9N2), have been observed to cause human
infections. The most notable of these is the A (H5N1) subtype which has become
established in domestic poultry throughout southeast Asia and has spread to
Europe and Africa in either wild birds or domestic poultry. Although growing
numbers of people have contracted the virus by contact with birds and there is
a high mortality rate (of 50%), there has been no evidence of ongoing person to
person transmission.

Clinical features
Influenza is transmitted from person to person via virus-containing respiratory
aerosols, droplets produced during coughing or sneezing, or by direct contact
with respiratory secretions.1,2 Influenza virus infection causes a wide spectrum
of disease from minimal or no symptoms, to respiratory illness with systemic
features, to multisystem complications and death from primary viral or

184 The Australian Immunisation Handbook 9th Edition

In adults, the onset of illness due to influenza is usually abrupt, after an


incubation period of 1 to 3days, and includes systemic features such as malaise,
feverishness, chills, headache, anorexia, and myalgia. These may be accompanied
by a cough, nasal discharge and sneezing. Fever is a prominent sign of infection
and peaks at the height of the systemic illness. Symptoms are similar for
influenza A and B viruses. However, infections due to influenza A (H3N2)
strains are more likely to lead to severe morbidity and increased mortality than
influenza B or influenza A (H1N1) strains.1,2
The clinical features of influenza A in infants and children are similar to those
in adults. However, temperatures may be higher in children (and may result
in febrile convulsions in the susceptible age group) and otitis media and
gastrointestinal manifestations are more prominent. Infection in neonates may be
associated with more non-specific symptoms.
Complications of influenza include acute bronchitis, croup, acute otitis
media, pneumonia (both primary viral and secondary bacterial pneumonia),
cardiovascular complications including myocarditis and pericarditis,
post-infectious encephalitis, Reye syndrome, and various haematological
abnormalities. Primary viral pneumonia occurs rarely, but secondary bacterial
pneumonia is a frequent complication in individuals whose medical condition
makes them vulnerable to the disease. Such individuals are at high risk in
epidemics and may die of pneumonia or cardiac decompensation.

Epidemiology
In mostyears, minor or major epidemics of type A or type B influenza
occur, usually during the wintermonths. In Australia, 85 deaths and 4250
hospitalisations are notified, on average, per year, although this is almost
certainly an underestimate due to failure to recognise the excess mortality
and hospitalisation associated with the disease. Extrapolation from US
estimates, based on more detailed surveillance, suggests 2000 deaths and 10000
hospitalisations are likely to occur annually in Australia. During epidemics, the
mortality rises, especially among the elderly and people with chronic diseases,
and there is increased morbidity and hospitalisation for pneumonia and
exacerbation of chronic diseases.3
Figure 3.9.1 shows the Australian hospitalisation and notification data for the
period 20032005.

Influenza 185

3.9 Influenza

secondary bacterial pneumonia. Severe disease is more likely with advanced


age, lack of previous exposure to antigenically related influenza virus, greater
virulence of the viral strain, chronic conditions such as heart or lung disease,
renal failure and diabetes, chronic neurological conditions, pregnancy, and
smoking. Annual attack rates in the general community are typically 5 to 10%,
but may be up to 20% in someyears. In households and closed populations,
attack rates may be 2 to 3 times higher.2

Figure 3.9.1: Influenza notification rates 20032005 and hospitalisation rates


2002/2003 to 2004/2005, Australia,* by age group4

Rate per 100,000

90

80

Rate per 100,000 population

70

Notifications
Hospitalisations

60

200
180
160
140
120
100
80
60
40
20
0
0

Age (years)

50

40

30

20

10

0
0-4

5-9

10-14

15-19

20-24

25-29

30-34

35-39

40-44

45-49

50-54

55-59

60-64

65-69

70-74

75-79

80-84

>=85

Age (years)

* Notifications where themonth of diagnosis was between January 2003 and December 2005;
hospitalisations where themonth of separation was between 1 July 2002 and 30 June 2005.

Pandemic influenza
It is now recognised that influenza A viruses have evolved in birds and that all
16 subtypes of influenza A persist in the avian reservoir. Occasionally, human
infections may occur, with influenza subtypes not currently present in the human
population, through close contact with infected poultry or poultry products.
These may result, as in the case of recent A (H5N1) infections, in severe or fatal
disease.
Avian influenza viruses are not naturally transmissible from person to person.
However, adaptation to human to human transmission can occur either if an
individual is concurrently infected with a human and an avian influenza virus,
permitting genetic reassortment to occur, or if the virus acquires this ability
via mutation. Genetic studies have shown that avian influenza viruses are the
source of new human pandemic strains and that both these processes resulted in
pandemic influenza in the 20th century.
Vaccines in routine inter-pandemic use will not protect against a pandemic strain
which, by definition, is new and unpredictable. If a pandemic occurs, there
will be a delay in producing a pandemic vaccine. Once the pandemic vaccine
is available, the priority groups and the timing of vaccination may be quite
different from those during inter-pandemic periods. In addition, the number of
vaccine doses required to confer protection and the optimal interval between
doses may differ. The Australian Influenza Pandemic Planning Committee has

186 The Australian Immunisation Handbook 9th Edition

developed guidelines for vaccine use and will advise health authorities about
priority groups, dosing schedules and timing of vaccination, should a pandemic
occur.

Vaccines
The administration of influenza vaccine to individuals at risk of complications
of infection is the single most important measure in preventing or attenuating
influenza infection and preventing mortality. After vaccination, most adults
develop antibody titres that are likely to protect them against the strains of virus
represented in the vaccine. In addition, the individual is protected against many
related variants. Infants, the very elderly, and patients with impaired immunity
may develop lower post-vaccination antibody titres. Under these circumstances,
influenza vaccine may be more effective in preventing lower respiratory tract
involvement or other complications of influenza than in preventing infection.
Fluad Delpharm Consultants/Novartis Vaccines (inactivated influenza
vaccine). Each 0.5mL pre-filled syringe contains 15g haemagglutinin of the
3 recommended strains, adjuvanted with MF59C; 0.05mg thiomersal. May
contain traces of kanamycin, neomycin, formaldehyde and egg protein.
Fluarix GlaxoSmithKline (inactivated influenza vaccine). Each 0.5mL prefilled syringe contains 15g haemagglutinin of each of the 3 recommended
strains; polysorbate 80/octoxinol 9. May contain traces of thiomersal,
formaldehyde, gentamicin and egg protein.
Fluvax CSL Biotherapies (inactivated influenza vaccine). Each 0.5mL prefilled syringe contains 15g haemagglutinin of each of the 3 recommended
strains. May contain traces of neomycin, polymyxin and egg protein.
Fluvirin Medeva/Ebos Health & Science (inactivated influenza vaccine).
Each 0.5mL pre-filled syringe contains 15g haemagglutinin of the 3
recommended strains. May contain traces of neomycin, polymyxin and egg
protein.
Influvac Solvay Pharmaceuticals (inactivated influenza vaccine). Each
0.5mL pre-filled syringe contains 15g haemagglutinin of each of the 3
recommended strains. May contain traces of gentamicin and egg protein.
Vaxigrip Sanofi Pasteur Pty Ltd (inactivated influenza vaccine). Each
0.5mL pre-filled syringe contains 15g haemagglutinin of each of the 3
recommended strains. May contain traces of formaldehyde, neomycin and
egg protein.

Influenza 187

3.9 Influenza

See http://www.health.gov.au/internet/wcms/publishing.nsf/Content/phdpandemic-plan.htm.

Vaxigrip Junior Sanofi Pasteur Pty Ltd (inactivated influenza vaccine).


Each 0.25mL pre-filled syringe contains 7.5g haemagglutinin of each of
the 3 recommended strains. May contain traces of formaldehyde, neomycin
and egg protein.
Fluvax and Fluarix each have a marking on the syringe to allow preparation of a
0.25mL dose suitable for paediatric use.
All the influenza vaccines currently available in Australia are either split virion
or subunit vaccines prepared from purified inactivated influenza virus which
has been cultivated in embryonated hens eggs. Split virion and subunit vaccines
are generally considered to be equivalent with respect to safety and efficacy, and
both are substantially free of the systemic reactions sometimes induced by whole
virus vaccines. Because the vaccine viruses are cultivated in embryonated hens
eggs, the vaccine may contain traces of egg-derived proteins. Manufacturing
processes vary by manufacturer, and different chemicals (formaldehyde or
betapropiolactone) may be used to inactivate the virus. Some influenza vaccines
distributed in Australia contain thiomersal, a mercury-containing compound,
as preservative, and other antibacterials or antibiotics may be used in the
manufacturing process. The product information should be consulted for specific
information.
Influenza vaccines normally contain the 3 recommended strains of virus, 2
current influenza A subtypes and influenza B, representing recently circulating
viruses. The final product contains 15g of viral haemagglutinin, the principal
surface antigen, for each virus strain. Vaxigrip Junior contains 7.5g of viral
haemagglutinin of each of the 3 recommended strains found in the adult
formulations. The composition of vaccines for use in Australia is determined
annually by the Australian Influenza Vaccine Committee.
Other forms of influenza vaccines (such as live attenuated intranasal vaccine)
have not yet been licensed in Australia.5
The effectiveness of influenza vaccine depends primarily on the age and
immunocompetence of the vaccine recipient and the degree of similarity between
the virus strains in the vaccine and those circulating in the community. In healthy
individuals <65years of age, influenza vaccine is 70 to 90% effective when the
antigenic match between vaccine and circulating viruses is close.6 Among elderly
people, the vaccine is 30 to 70% effective in preventing all hospitalisation for
pneumonia and influenza for those living outside nursing homes or similar
chronic-care facilities. For those residing in nursing homes, influenza vaccine
is most effective in preventing severe illness, secondary complications and
deaths. In such a population, the vaccine can be 50 to 60% effective in preventing
hospitalisation or pneumonia, and 80% effective in preventing death, even
though the effectiveness in preventing influenza illness may be lower.7 Currently

188 The Australian Immunisation Handbook 9th Edition

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.8 Store
at +2C to +8C. Do not freeze. At the end of each year, vaccine should be
appropriately discarded to avoid inadvertently using a product with incorrect
formulation in the following year.

Dosage and administration


Shake the pre-filled syringe vigorously before injection. Influenza vaccine is
administered by either IM or SC injection. The IM route causes fewer local
reactions and is preferred.9
Table 3.9.1: Recommended doses of influenza vaccine
Age

Dose

Number of doses
(first vaccination)

Number of doses*
(subsequentyears)

6months<3years

0.25mL

39years

0.5mL

>9years

0.5mL

* If a child 6months to 9years of age receiving influenza vaccine for the first time
inadvertently does not receive the second dose within the same year, he/she should have 2
doses administered the following year.7
Two doses at least 1month apart are recommended for children aged 9years who are
receiving influenza vaccine for the first time. The same vial should not be re-used for the 2
doses.

Note:
(i) Some influenza vaccines available in Australia are packed in syringes
graduated for measurement of recommended paediatric doses. Vaxigrip Junior
presentation is a 0.25mL pre-filled syringe ready for use. Fluvax and Fluarix
each have a marking on the syringe to allow preparation of a 0.25mL dose.
A tuberculin syringe can be used to measure the dose of vaccine not packed
in graduated syringes. Excess vaccine is expelled from the syringe and the
remaining volume injected.
(ii) All the product information sheets have some differences from Table 3.9.1.
Fluvirin does not have a dose recommendation for children <4years of age. The
safety of Fluad, which is adjuvanted with MF59C, has not been established in
children and it is registered for use only in people 65years of age.

Influenza 189

3.9 Influenza

available influenza vaccines confer protection for about a year. Low levels of
protection may persist for a further year, if the prevalent strain remains the
same or undergoes only minor antigenic drift. To provide continuing protection,
annual vaccination with vaccine containing the most recent strains is necessary.

Vaccination is best undertaken in autumn, in anticipation of winter outbreaks of


influenza. However, vaccination can be given as early as February. In autumn,
the opportunities to provide influenza vaccination to individuals at increased
risk should not be missed when they present for routine care.
As full protection is usually achieved within 10 to 14days and there is evidence
of increased immunity within a fewdays, vaccination can still be offered to
adults and children after influenza virus activity has been documented in a
community.
Influenza vaccine can be administered concurrently with other vaccines,
including pneumococcal polysaccharide vaccine and all the scheduled childhood
vaccines.

Recommendations
Annual influenza vaccination is recommended for any person 6months of age
who wishes to reduce the likelihood of becoming ill with influenza.
Influenza vaccination is strongly recommended and should be actively promoted
for the following groups:

1. People at increased risk of complications from influenza infection


(i) All individuals 65years of age3,10-13
Influenza vaccine has been shown to reduce hospitalisations from pneumonia
and all-cause mortality by about half in adults 65years of age.

(ii) All Aboriginal and Torres Strait Islander people 15years of age
Annual influenza vaccine is recommended for Aboriginal and Torres Strait
Islander people 15years of age in view of the substantially increased risk of
hospitalisation and death from influenza and pneumonia (see Chapter 3.15,
Pneumococcal disease). In Aboriginal and Torres Strait Islander people 50years of
age, routine pneumococcal polysaccharide vaccination is also recommended (see
Chapter 2.1, Vaccination for Aboriginal and Torres Strait Islander people and Chapter
3.15, Pneumococcal disease).

(iii) Individuals 6months of age with conditions


predisposing to severe influenza
Cardiac disease including cyanotic congenital heart disease, coronary artery
disease and congestive heart failure.14,15 Influenza causes increased morbidity
and mortality in children with congenital heart disease and adults with
coronary artery disease and congestive heart failure.14
Chronic respiratory conditions including:
Suppurative lung disease, bronchiectasis, and cystic fibrosis.16 Patients
with these diseases are at greatly increased risk from influenza, which
may cause irreversible deterioration in lung function.

190 The Australian Immunisation Handbook 9th Edition

Severe asthma. In patients with severe asthma, defined as requiring


frequent hospital visits, annual influenza vaccine is an important part of
routine care.17-19 There are insufficient data from randomised controlled
trials of influenza vaccine to define efficacy across the whole spectrum
of asthma,20 but influenza can cause severe exacerbations of wheezing,
and about 10% of episodes of virus-induced wheezing are attributable to
influenza.
Other chronic illnesses requiring regular medical follow-up or hospitalisation in the
preceding year, including:
diabetes mellitus,
chronic metabolic diseases,
chronic renal failure,
haemoglobinopathies, and
impaired immunity (including drug-induced immune impairment).7,21,22
Chronic neurological conditions (eg. multiple sclerosis, spinal cord injuries,
seizure disorders or other neuromuscular disorders) that can compromise
respiratory function or the expulsion of respiratory secretions or that can
increase the risk for aspiration.7 NB. Because they can experience severe, even
fatal, influenza, vaccination is particularly important for children 6months
of age with chronic neurological conditions.7
People with impaired immunity, including HIV infection.23,24 Patients with
impaired immunity, including HIV infection, malignancy and chronic steroid
use, are at greatly increased risk from influenza, although they also have
a reduced immune response to the vaccine. While patients with advanced
HIV disease and low CD4 T-lymphocyte counts may not develop protective
antibody titres, there is evidence that for those with minimal symptoms
and high CD4 T-lymphocyte counts (see Chapter 2.3, Groups with special
vaccination requirements, Table 2.3.4), protective antibody titres are obtained
after influenza vaccination.24 Influenza vaccine has been shown in a clinical
trial to reduce the incidence of influenza in HIV-infected patients,24 and
although viral load may increase transiently, there is no impact on CD4
count.23
Long-term aspirin therapy in children (aged 6months to 10years). Such children
are at increased risk of Reye syndrome after influenza.

Influenza 191

3.9 Influenza

Chronic obstructive pulmonary disease and chronic emphysema. There


is clinical trial evidence that inactivated influenza vaccination has a
clinically important protective effect on influenza-related exacerbations,
and probably an effect on the total of exacerbations in COPD patients.
There is no evidence that inactivated influenza vaccination causes
exacerbations of COPD.16

(iv) Pregnant women


It is recommended that influenza vaccine be offered in advance to women
planning a pregnancy, and to pregnant women who will be in the second or third
trimester during the influenza season, including those in the first trimester at
the time of vaccination.25,26 Influenza vaccination is estimated to prevent 1 to 2
hospitalisations per 1000 women vaccinated during the second or third trimester.

(v) Residents of nursing homes and other long-term care facilities


This is due to high rates of transmission and complications during outbreaks.3,9-13,27

(vi) Homeless people and those providing care to them


The living conditions and prevalence of underlying medical conditions among
homeless people will predispose to complications and transmission of influenza.

2. People who may potentially transmit influenza to


those at high risk of complications from influenza
The following groups of people can potentially transmit influenza to high-risk
patients and it has been shown that vaccinating the former protects those at
high-risk:
staff of nursing homes,
healthcare providers28 (particularly of patients with impaired immunity),
staff of long-term care facilities,
household contacts (including children 6months of age) of individuals in
high-risk groups.

3. People involved in the commercial poultry industry or in


culling poultry during confirmed avian influenza activity29
Vaccination using the current influenza season vaccine composition is
recommended for poultry workers and others in regular close contact with
poultry during an avian influenza outbreak.29 Although routine influenza vaccine
does not protect against avian influenza, there is a possibility that a person who
was infected at the same time with avian and human strains of influenza virus
could allow reassortment of the 2 strains to form a virulent strain that could
spread from human to human (ie. initiate a pandemic).

4. People providing essential services


Vaccination of those who provide essential community services will minimise
disruption of essential activities during influenza outbreaks. Influenza viral
infections can place considerable pressure upon both public and private
healthcare services (see Chapter 2.3, Groups with special vaccination requirements,
Table 2.3.6).

192 The Australian Immunisation Handbook 9th Edition

5. Workers in other industries

6. Travellers
Large tourist groups, especially those including elderly people and those
travelling on cruises, who are likely to be in confined circumstances fordays
toweeks, are at risk of influenza, either acquired before departure or from
travel to areas of the world where influenza is currently circulating. Influenza
vaccination, preferably using the strain prevalent in the areas in which they
will be travelling, is recommended if travelling during the influenza season,
especially if it is known before travel that there are high rates or epidemics of
influenza.7

Contraindications
Absolute contraindications to influenza vaccine are:
anaphylaxis following a previous dose of any influenza vaccine,
anaphylaxis following any vaccine component,
individuals with anaphylactic sensitivity to eggs should not be given
influenza vaccine. This includes those who, soon after ingesting eggs,
develop swelling of the lips or tongue, or experience acute respiratory
distress or collapse.

Precautions
Patients with a history of Guillain-Barr Syndrome (GBS) with an onset related in
time to influenza vaccination may be at increased risk of again developing GBS
if given influenza vaccine. The risk should be weighed against the benefits to the
individual patient of influenza vaccination. Because patients with a history of
GBS have an increased likelihood of developing the syndrome again, the chance
of them coincidentally developing the syndrome following influenza vaccination
may be higher than in individuals with no history of GBS.

Adverse events27,30
Local adverse events (induration, swelling, redness and pain) are very common
(>10%).
Fever, malaise and myalgia occur commonly (110%). These adverse events may
commence within a few hours of vaccination and may last for 1 to 2days. In
children <5years of age, these adverse events may be more pronounced. Postvaccination symptoms may mimic influenza infection but current influenza
vaccines do not contain live virus and cannot cause influenza.

Influenza 193

3.9 Influenza

The cost-effectiveness of influenza vaccination in industry varies from year to


year, depending on the amount of circulating influenza, but the overall impact
over time is judged to be cost-saving in several settings.6,7 Individual industries
should consider the benefits of offering influenza vaccine in the workplace.

Immediate adverse events (such as hives, angioedema, or anaphylaxis) are a


rare consequence of influenza vaccination. They probably represent an allergic
response to a residual component of the manufacturing process, most likely egg
protein. Individuals with a history of anaphylaxis after eating eggs or a history of
a severe allergic reaction following occupational exposure to egg protein should
not be given influenza vaccine.
An association was shown between influenza vaccine used in the northern
hemisphere from 1992 to 1994 and Guillain-Barr syndrome (GBS), with 1 to
2 cases of GBS occurring per million vaccinated. There has not been an excess
number of cases of GBS reported in Australia in association with influenza
vaccine.31

Use in pregnancy
Influenza vaccine is recommended for pregnant women who will be in the
second or third trimester during the influenza season, including those in the first
trimester at the time of vaccination. See Recommendations above.

Variations from product information


The product information lists allergy to chicken feathers and some food proteins
as a contraindication, whereas NHMRC recommends that patients with allergies
other than anaphylaxis can be vaccinated.
The product information for Fluarix states the influenza vaccine may be used
in children from 3months of age. NHMRC recommends influenza vaccine in
children 6months of age.
The product information for some vaccines gives a dose of 0.125mL for children
3 or 6months to 2years old. NHMRC recommends that the lowest dose for any
influenza vaccine is 0.25mL. This is because influenza vaccine is relatively poorly
immunogenic in infants, and 0.25mL is the dose recommended in the USA for
children aged 6months where it has been shown to be safe.32
The product information for Fluvirin states that the product should not be given
to children <4years of age. Although the NHMRC recommends that children as
young as 6months of age can be vaccinated if they are at risk of complications of
influenza, the suitability of the vaccine formulation for accurate preparation of
0.25mL doses should be taken into account.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

194 The Australian Immunisation Handbook 9th Edition

3.10 Japanese encephalitis


Virology
Japanese encephalitis (JE) is caused by a mosquito-borne flavivirus.

Clinical features

Epidemiology
JE is a significant public health problem in many parts of Asia including the
Indian subcontinent, southeast Asia and China.1 In recentyears, however, the
disease has extended beyond its traditionally recognised boundaries with, for
example, outbreaks occurring in the Torres Strait and north Queensland in 1995
and 1998.2,3
The JE virus is essentially a zoonosis of pigs and wading birds, and is transmitted
between these animals by Culicine mosquitoes.1 The virus replicates, leading to
a transient high-level viraemia, within these so-called amplifying hosts but not
within other large vertebrates such as horses and humans.
Indeed, humans are an incidental host infected when living in close proximity
to the enzootic cycle; this usually occurs in rural areas where there is prolific
breeding of the vectors in flooded rice fields.1
There are two recognised epidemiological patterns of JE.1 In the temperate or
subtropical regions of Asia (northern Thailand, northern Vietnam, Korea, Japan,
Taiwan, China, Nepal and northern India), the disease occurs in epidemics during
the summer or wet seasonmonths (April to May until September to October). In
the tropical regions (most of southeast Asia, Sri Lanka, southern India), the disease
is endemic, occurring throughout the year, but particularly during the wet season.1
In some countries (Japan, Taiwan, South Korea, and some provinces of China),
the incidence of JE has declined considerably in recent decades, and it has been
eradicated from Singapore. Immunisation, changes in pig husbandry, a reduction
in land utilised for rice farming, and improved socioeconomic circumstances
have all contributed to these changes.1

Japanese encephalitis 195

3.10 Japanese encephalitis

The disease is typically an acute neurological illness characterised by headache,


fever, convulsions, focal neurological signs and depressed level of consciousness.
It has a high case-fatality rate and there is a high prevalence of neurological
sequelae (up to 50%) in those who survive the acute illness.1 Less commonly, the
disease may present as an acute flaccid paralysis.1 Milder forms include febrile
illness with headache, and aseptic meningitis. It is recognised, however, that
most infections are asymptomatic; published estimates of the symptomatic to
asymptomatic infection ratio vary in different populations from 1:25 to 1:1000.1

In early 1995, 3 cases of JE, 2 of them fatal, occurred on Badu island in the
Torres Strait.2 Subsequent serological surveys showed that JE virus activity was
widespread in many other remote outer islands of the Torres Strait (see Figure
3.10.1) at or about that time.2 Although the 1995 outbreak was the first known
incursion of JE virus into Australia, surveillance using sentinel pigs has shown
incursions into the Torres Strait in virtually every wet season (December to May)
since then.
In early 1998, a further case of JE occurred in an unvaccinated Badu resident and
the first ever mainland case of JE occurred in a person working on the west coast
of Cape York.3 However, serological surveys revealed no evidence of JE virus
infection in people in several nearby communities.3 To date, there have been 5
cases of JE acquired in Australia.
An investigation subsequent to the 1995 outbreak of JE in the Torres Strait
documented the presence of the JE virus in the Western Province of Papua New
Guinea.4 A severe case of JE acquired near Port Moresby occurred in early 2004,5
indicating that the JE virus is now probably widespread in Papua New Guinea.
Figure 3.10.1: Map of the Torres Strait. The outer islands are north of the dotted
line

Vaccine
JE-VAX Sanofi Pasteur Pty Ltd (Japanese encephalitis virus vaccine
inactivated). Each 1.0mL reconstituted monodose vial contains
formaldehyde inactivated Japanese encephalitis virus; 0.007% thiomersal;
470g gelatin; <100g formaldehyde; 5mg monosodium glutamate; <50
ng mouse brain serum protein.

196 The Australian Immunisation Handbook 9th Edition

The JE vaccine available in Australia is an inactivated mouse brain-derived


vaccine manufactured in Japan. However, the manufacturer has recently
discontinued its production and, although the Australian distributor has access
to a stockpile, shortages of the vaccine could occur over the next fewyears. New
generation JE vaccines are expected in the mid to longer term.
The vaccine is prepared by inoculating mice intracerebrally with NakayamaNIH strain JE virus. Infected brains are harvested, homogenised, then
centrifuged. The supernatant is inactivated with formaldehyde and purified by
ultracentrifugation; the suspension is then lyophilised. No myelin basic protein
can be detected at the threshold of the assay (<2 ng/mL).

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.8 Store at
+2C to +8C. Do not freeze. Reconstituted vaccine should be used immediately,
but it can be stored at +2C to +8C and used within 8 hours.

Dosage and administration


JE vaccine is administered by the subcutaneous route. The volume injected is
0.5mL in 13-year-old children and 1.0mL for all individuals >3years of age. In
those from non-endemic regions, including Australia, a 3-dose regimen (ie.days
0, 7 and 28) over amonth is required. An accelerated schedule of 0, 7 and 14days
can be used, but this is likely to result in lower antibody levels than the standard
schedule.7 If the accelerated schedule is used, a further dose should, if possible,
be administered 1 to 3months later.
NB. The volume of the reconstituted vaccine is greater than 1.0mL. Because the
dose of JE vaccine is 1.0mL (0.5mL in 13-year-old children) this means a small
portion of the total reconstituted vaccine should be discarded.

Recommendations
(i) Travellers
Although the risk of travellers in Asia acquiring JE is extremely low, there have
been instances of even short-term travellers developing the disease.9 Therefore, all
travellers to Asia (and other tropical regions) must be fully aware of the need to
take appropriate measures to avoid mosquito bites.
The risk of JE to travellers to Asia is determined by the season of travel, the
regions visited, the duration of travel, the extent of outdoors activity and the
extent to which mosquito-avoidance measures are taken.1 Clearly the risk is

Japanese encephalitis 197

3.10 Japanese encephalitis

A randomised clinical trial in Thailand in the early 1980s determined that 2


doses of the inactivated mouse brain-derived vaccine, administered to children
7days apart, had a protective efficacy of 91%.6 However, immunogenicity studies
have demonstrated that 3 doses of the vaccine are required to ensure adequate
immunity in vaccinees from JE non-endemic areas.7

greater during prolonged travel to rural areas of Asia during the wet season; it is
probably negligible during short business trips to urban areas.
NB. A recent study has shown that the JE virus is hyperendemic in Bali, that it
causes substantial human illness, and that it circulates year round.10
Therefore JE vaccination is recommended for:
travellers spending 1month or more in rural areas of Asia. However, as
JE has occurred in travellers after shorter periods of travel, JE vaccination
should be considered for shorter-term travellers, particularly if the travel is
during the wet season, and/or there is considerable outdoor activity, and/or
the accommodation is not mosquito-proof,9
for all other travellers spending a year or more in Asia (except Singapore),
even if much of the stay is in urban areas, and
travellers intending to spend amonth or more in Papua New Guinea,
particularly if the travel is during the wet season.

(ii) Residents of Far North Queensland


JE vaccination is recommended for:
all residents (>1 year of age) of the outer islands in the Torres Strait, and
all non-residents who will be living or working on the outer islands of the
Torres Strait for a cumulative total of 30days or more during the wet season
(December to May).
NB. The period of greatest risk is from February to March and the vaccination
course should be completed before February. Those arriving in the outer islands
late in the wet season (ie. in May) have arrived after the risk period and do not
require vaccination. Those visiting the outer islands in the dry season (June to
November) do not require vaccination. Those visiting only the inner islands,
including Thursday Island, do not require vaccination.

(iii) Laboratory personnel


JE vaccination is recommended for all research laboratory personnel who
potentially might be exposed to the virus.

(iv) Booster doses


Single booster doses are recommended at 3-yearly intervals.

198 The Australian Immunisation Handbook 9th Edition

Contraindications
Anaphylaxis following a previous dose of JE vaccine or a significant allergic
reaction, such as generalised urticaria, to a previous dose.
Anaphylaxis following any component of the vaccine. A past history
of allergic disorders (including urticaria, angioedema, anaphylaxis)
following bee-stings, medications, foods etc. must be considered as possible
contraindications to vaccination.
The inactivated mouse brain-derived JE vaccine is contraindicated in those <1
year of age.

There are few data on the safety and efficacy of JE vaccine in people with impaired
immunity. A small study undertaken in Thailand has documented that HIVinfected infants respond less well to 2 doses of JE vaccine than do non-infected
infants;11 the response to further doses was not studied.

Adverse events
Local reactions and minor systemic reactions are common to very common
following vaccination against JE.1 About 20% experience tenderness, redness
and/or swelling at the injection site, and 10% experience systemic reactions
such as fever, headache, being off-colour, chills, dizziness, aching muscles,
nausea and/or vomiting.
Although the manufacturing process purifies the infected mouse brain
suspension so that no myelin basic protein can be detected in the vaccine,
serious neurological events following immunisation have been reported. In
1994, 4 cases of severe neurological illness, 2 of which were fatal, were reported
from South Korea, and surveillance in Japan indicates the rate of severe
neurological adverse events following JE vaccination is 1.8 cases per 1 million
doses of vaccine.7
Hypersensitivity (allergic) reactions are uncommon and occur in about
0.5% (ie. 1 in 200) vaccinees. These reactions include urticaria that is often
widely distributed over the body, angioedema of the limbs, face and throat,
and generalised pruritus (sometimes without a rash). In the early 1990s,
apparently severe allergic reactions to the inactivated mouse brain-derived
JE vaccine were reported from several industrialised countries, including
Australia.7 In a few cases, upper airway swelling with respiratory distress
and hypotension occurred; some had to be hospitalised.
An important feature of the hypersensitivity reactions to JE vaccine is that they
may be delayed for severaldays, in some cases up to 10days, after the actual
time of vaccine administration. The risk of these delayed reactions seems to be
increased after the first and second doses, and they appear to be more likely to
occur in those with a history of allergic reactions, especially urticaria.7 Although

Japanese encephalitis 199

3.10 Japanese encephalitis

Precautions

the pathogenesis of the more severe hypersensitivity reactions remains uncertain,


there is some evidence that gelatin, added to stabilise the vaccine, may be the
provoking agent.7 As a precaution, vaccinees should ideally remain within access
to medical care for 10days after vaccination.

Use in pregnancy
Although JE vaccine might pose a theoretical risk to the developing fetus, no
adverse outcomes of pregnancy have ever been attributed to vaccination against
JE. Because JE virus infection during the first and second trimester is also
associated with miscarriage, pregnant women at risk of acquiring JE should be
offered JE vaccine.

Variations from product information


The product information states that definitive recommendations cannot be given
on the timing of booster doses at this time and that a booster dose may be given
after 2years. The NHMRC recommends that single booster doses be given at
3-yearly intervals.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

200 The Australian Immunisation Handbook 9th Edition

3.11 Measles
Virology
Measles is a paramyxovirus, genus Morbillivirus. It is an RNA virus with 6
structural proteins, 3 complexed to the RNA and 3 associated with the viral
envelope. Two of the envelope proteins, the F (fusion) protein and the H
(haemagglutinin) protein, are the most important in pathogenesis. The measles
virus can survive up to 2 hours in air, but is rapidly inactivated by heat, light and
extremes of pH.1,2

Clinical features
Measles is a highly infectious, acute viral illness spread by respiratory secretions,
including air-borne transmission via aerosolised droplets.2 It is infectious from
the beginning of the prodromal period and up to 4days after the appearance of
the rash. The incubation period is usually 10 to 14days. The prodrome, lasting 2
to 4days, is characterised by fever and malaise followed by a cough, coryza and
conjunctivitis. The maculopapular rash typically begins on the face and upper
neck, and then becomes generalised.

Epidemiology
Evidence suggests that endemic measles has been eliminated from Australia,
an indigenous measles strain being absent for severalyears.4 Although measles
outbreaks of limited duration continue to occur, they have usually been linked
to imported cases.5-7 In a recent measles outbreak, linked to an imported case,
25% of notified cases were in children aged 14years, most of whom were not
vaccinated.8 Measles notifications and hospitalisations for the 5years 20012005
have been the lowest recorded in Australia.9,10 In the 30years (19762005) since
measles vaccination was recommended in Australia, there have been 95 deaths

Measles 201

3.11 Measles

Measles is often a severe disease, frequently complicated by otitis media


(in ~9%), pneumonia (in ~6%) and diarrhoea (in ~8%).1,2 Acute encephalitis
occurs in 1 per 1000 cases, and has a mortality rate of 10 to 15%, with 15 to
40% of survivors suffering permanent brain damage.3 Subacute sclerosing
panencephalitis (SSPE) is a late complication of measles, occurring on average
7years after infection in approximately 0.5 to 1 per 100000 measles cases.2
SSPE causes progressive brain damage and is always fatal. Complications from
measles are more common and more severe in the chronically ill, in children
<5years of age, and in adults.1 Approximately 60% of deaths from measles are
attributed to pneumonia, especially in the young, while complications from
encephalitis are more frequently seen in adults.1,2 Measles infection during
pregnancy can result in miscarriage and premature delivery but has not been
associated with congenital malformation.1

recorded from measles, 1 death in 2004 being the only one recorded since 1995.9-11
High-level vaccination coverage is imperative to maintain measles elimination,
requiring rates for each new birth cohort of >95% for a single dose and >90%
for 2 doses.12 In 2004, the Australian Childhood Immunisation Register (ACIR)
recorded that 93.6% of children aged 2years (born in 2002) had received at least
1 dose of measles-containing vaccine and 84.8% of children aged 6years (born
in 1998) had received both doses.13 It is likely that, when corrected for underreporting, the target of 95% coverage for 1 dose of measles vaccine is reached
at 2years, but, if the second dose is not given until 4years of age, 95% 2-dose
coverage is not achieved.14 Scheduling of the second dose of measles-containing
vaccine at 18months of age (see Recommendations below) will provide 2-dose
protection at an earlier age and may also improve second dose coverage.
Following the National Measles Control Campaign (which took place in 1998
and resulted in 1.7 million primary school children being vaccinated), a national
serosurvey in the first quarter of 1999 showed that 89% of children aged
25years, 94% of those aged 611years, and 91% of those aged 1218years,
were immune to measles.15,16 The serosurvey evaluating the young adult MMR
campaign in 2000 showed that those most at risk of measles infection in Australia
were infants <12months of age, 1<2-year-olds due to delayed vaccine uptake,
and individuals born in the late 1960s to mid 1980s (especially the 19781982
birth cohort).17 Young adults are recognised to be at a greater risk of measles
infection. Many missed being vaccinated as infants (when coverage was low),
while during their childhood a second dose was not yet recommended and
disease exposure was decreasing.18
Worldwide, measles is thought to be the fifth leading cause of childhood
morbidity and mortality with 770000 deaths estimated to have occurred in
2000. More than half these deaths occurred in Africa.1,19 Following extensive
vaccination campaigns, measles accounted for approximately 454000 deaths
worldwide in 2004.20 The WHO is overseeing efforts to eliminate measles
worldwide through immunisation and surveillance strategies that aim to
interrupt the circulation of the virus.21

Vaccines
One measles-mumps-rubella (MMR) vaccine is currently available in Australia.
It is anticipated that measles-mumps-rubella-varicella (MMRV) vaccines will
become available in the near future. A monovalent vaccine is available for
rubella where this is specifically required (see Chapter 3.19, Rubella). Separate
administration of measles, mumps or rubella vaccine is not recommended as an
alternative to MMR vaccine and no monovalent vaccines for mumps or measles
are licensed in Australia.
Measles immunity induced by single-dose vaccination provides long-term
immunity in most recipients.1,22 However, approximately 5% of recipients fail to
develop immunity to measles after 1 dose.23 Following a second vaccine dose,

202 The Australian Immunisation Handbook 9th Edition

approximately 99% of subjects overall will be immune to measles. Combination


MMRV vaccines have been shown, in clinical trials, to produce similar rates
of seroconversion to all 4 vaccine components compared with MMR and
monovalent varicella vaccines administered at separate injection sites.24,25 Data on
the use of MMRV vaccines are not available for people >12years of age.
Priorix (MMR) GlaxoSmithKline (live attenuated measles virus (Schwarz
strain), RIT 4385 strain of mumps virus (derived from the Jeryl Lynn strain)
and the Wistar RA 27/3 rubella virus strain). Each 0.5mL monodose of the
reconstituted, lyophilised vaccine contains not less than 103.0 CCID50 (cell
culture infectious dose 50%) of the Schwarz measles, not less than 103.7
CCID50 of the RIT 4385 mumps and not less than 103.0 CCID50 of the Wistar
RA 27/3 rubella virus strains; lactose; neomycin; amino acids; sorbitol and
mannitol as stabilisers.

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.26 Store at
+2C to +8C. Protect from light. Do not freeze. Reconstituted vaccine should be
used immediately, but can be stored at +2C to +8C for up to 8 hours before use.

Dosage and administration


For both children and adults, the dose of MMR is 0.5mL, administered by either
SC or IM injection.

Recommendations
(i) Routine vaccination of children
Two doses of MMR are recommended for all children. The first dose should
be given at 12months of age and the second dose at 18months of age. The
minimum interval between doses is 4weeks.
When MMRV vaccines are available, the 12month and 18month doses may be
given as MMRV.
The scheduled age at administration of the second dose of measles-containing
vaccine has been moved from 4years of age to 18months of age to provide
earlier 2-dose protection against measles and to improve vaccine uptake
(see Epidemiology above). The second dose of measles-containing vaccine
at 18months of age can be given as either MMR or, when available, MMRV.
Receipt of 2 doses of varicella vaccine (VV) provides increased protection against

Measles 203

3.11 Measles

MMR can be given at the same time as other vaccines (including DTPa, hepatitis
B, MenCCV and varicella), using separate syringes and injection sites. If MMR is
not given simultaneously with other live viral parenteral vaccines (eg. varicella
vaccine), they should be given at least 4weeks apart (see Precautions below).27,28

varicella, and MMRV, when available, should be preferred over MMR for the
second dose of measles-containing vaccine at 18months of age. (For further
information, see also Chapter 3.24, Varicella.)

(ii) Vaccination of adults and adolescents


Those born before 1966:
No vaccination is required (unless serological evidence indicates otherwise) as
circulating virus and disease were prevalent before this time suggesting most
people would have acquired immunity from natural infection. However, recent
confirmed cases of measles have occurred in individuals born before 1966 and, if
doubt exists, it may be more expedient to offer vaccination than serological testing.8
Those born during or since 1966:
Infants 12months up to 18months of age should have documented evidence of
1 dose of MMR (or MMRV when available), or serological evidence of protection
for measles.
Those 18months of age should have documented evidence of 2 doses of MMR
(administered at least 4weeks apart with both doses administered at 12months
of age or over), or serological evidence of protection for measles, mumps and
rubella.
Catch-up vaccination of children who have not received MMR or MMRV at
18months of age should occur at the 4-year-old schedule point, until all the
relevant children have reached 4years of age. It is also acceptable to use MMRV
in place of MMR at the 4-year-old schedule point. This would have the added
benefit of providing a 2-dose VV schedule to an additional 2.5 birth cohorts of
infants who had received single-dose monovalent VV at 18months.
There are no increased adverse events from vaccinating those with pre-existing
immunity to 1 or more of the vaccine components.

(iii) Healthcare workers and those who work with children


All workers in these categories who were born during or since 1966 and are
non-immune or who have only received 1 dose of MMR, should be vaccinated
with MMR, and have documented evidence of 2 doses or serological evidence
of protection for measles, mumps and rubella (see Vaccination of adults and
adolescents above). (See also Section 2.3, Table 2.3.6 Recommended vaccinations for
those at risk of occupationally acquired vaccine-preventable diseases.)

(iv) Travellers
Those born during or since 1966 should be encouraged to complete the MMR
vaccination schedule (using MMR or MMRV, when appropriate) before
embarking on international travel if they do not have evidence of receipt of 2
doses of MMR (see Vaccination of adults and adolescents above).

204 The Australian Immunisation Handbook 9th Edition

Infants travelling to endemic countries may be vaccinated with MMR between 9


and 12months of age. In these cases, another dose of MMR (or MMRV) should be
given at 12months of age or 4weeks after the first dose, whichever is later. This
should be followed by the routine administration of the next dose of MMR or MMRV
at 18months of age. This is because maternal antibodies to measles are known
to persist in many infants until 11months of age and may interfere with active
immunisation before 12months of age1 (see Vaccination during an outbreak below).

Contraindications
If using MMRV vaccine, additional contraindications relating to the varicella
vaccine component are outlined in Chapter 3.24, Varicella.

(i) Allergy to vaccine components


Vaccination is contraindicated where there has been:
anaphylaxis following a previous dose of MMR or MMRV, or
anaphylaxis following any component of the vaccine.
Providers should consult the product information regarding vaccine components
when MMRV vaccines are available.

(ii) People with impaired immunity

those with primary or acquired cellular immunodeficiency states, including


impaired immunity due to HIV/AIDS or conditions in which normal
immunological mechanisms may be impaired. MMR (but not MMRV)
vaccine can be given to HIV-positive children who do not have impaired
immunity (see Precautions below),
those taking high-dose oral corticosteroids (in children equivalent to either
>2mg/kg per day prednisolone (20mg per day total) for >1week or
>1mg/kg per day for >4weeks) (see Section 2.3.3, Vaccination of individuals
with impaired immunity due to disease or treatment),
those receiving high-dose systemic immunosuppressive treatment, general
radiation or x-ray therapy,
those suffering from malignant conditions of the reticuloendothelial system
(such as lymphoma, leukaemia, Hodgkins disease).

Measles 205

3.11 Measles

Measles-, mumps-, rubella- and varicella-containing vaccines are contraindicated


in individuals with impaired immunity. In addition, there are no clinical
trials or post-licensure data to address the safety and immunogenicity of
MMRV in children or adults with impaired immunity. However, based on
recommendations for the component live attenuated vaccine viruses, both MMR
and MMRV are contraindicated in the following groups:

(iii) Recent administration of antibody-containing blood product


The expected immune response to measles, mumps, rubella and varicella
vaccination may be impaired after receipt of antibody-containing blood
products.23,27,29 The duration of interference with the response to measles
vaccination depends on the amount of immunoglobulin contained in each
product, and ranges from 3 to 11months.27 Vaccination with MMR or MMRV
should be delayed after administration of antibody-containing products
as indicated in Table 2.3.5 (see Section 2.3.5, Vaccination of patients following
receipt of other blood products including blood transfusions).
After vaccination with MMR or MMRV, immunoglobulin-containing
products should not be administered for 3weeks unless the benefits
exceed those of vaccination. If immunoglobulin-containing products are
administered within this interval, the vaccinee should either be revaccinated
later at the appropriate time following the product as indicated in Table 2.3.5,
or tested for immunity 6months later and then revaccinated if seronegative.
Blood transfusion with washed red blood cells is not a contraindication to
MMR or MMRV vaccinations.
Rh (D) immunoglobulin (anti-D) does not interfere with the antibody
response to MMR vaccine and may be given at the same time in different
sites with separate syringes or at any time in relation to each other.

(iv) Pregnant women


If MMR vaccines are given to women of child-bearing age, pregnancy should
be avoided for 28days30 (see Chapter 3.19, Rubella). Data on the use of MMRV
vaccines in individuals >12years of age are not available.

Precautions
MMR can be administered on the same day as other live viral parenteral
vaccines, such as monovalent varicella vaccine. However, if this is not
possible, MMR should be deferred for at least 4weeks after vaccination with
other live viral parenteral vaccines.
MMR can be given to asymptomatic or mildly symptomatic HIV-positive
individuals providing they do not have severely impaired immunity.23 (see
Section 2.3.3, Table 2.3.4, Immunological categories based on age-specific CD4
counts and percentage of total lymphocytes). This is because the risk posed by
measles infection is considered to be greater than the likelihood of adverse
events from vaccination.31 As there are no data available on the safety,
immunogenicity or efficacy of MMRV vaccine in HIV-infected children,
MMRV vaccine should not be administered as a substitute for MMR when
vaccinating these children.23,29

206 The Australian Immunisation Handbook 9th Edition

Children on daily doses of 2mg/kg per day of systemic corticosteroids


for <1week, and those on lower doses of 1mg/kg per day or alternate-day
regimens for longer periods, may be given live viral vaccines.
Children receiving >2mg/kg per day or 20mg per day in total of
prednisolone (or equivalent) for >14days can receive live viral vaccines after
corticosteroid therapy has been discontinued for at least 1month.31 Some
experts suggest withholding lower doses of steroids 2 to 3weeks before
vaccination with live viral vaccines if this is possible29,31 (see Section 2.3.3,
Vaccination of individuals with impaired immunity due to disease or treatment).
Use of salicylates (aspirin) is not recommended for 6weeks following
MMRV vaccination. This is because of the association between use of
salicylates during natural varicella infection and Reye syndrome (see Chapter
3.24, Varicella). There is no need to avoid salicylates (aspirin) after MMR
vaccination.
Thrombocytopenia is a rare adverse event following vaccination with
MMR.2,32,33 Authorities differ in their opinion about whether the risk
of vaccine-associated thrombocytopenia is increased in those who
have previously had immune thrombocytopenia.23,33 Post-marketing
experience of live MMR vaccine in the USA indicates that individuals with
current thrombocytopenia may develop more severe thrombocytopenia
after vaccination. Recent studies found that children with immune
thrombocytopenia before MMR had no vaccine-associated recurrences.32,33
There are no systematic studies of the outcome of a second dose of MMR in
children who developed thrombocytopenia after a first dose.33

Measles virus inhibits the response to tuberculin, so tuberculin-positive


individuals may become tuberculin-negative for up to amonth after measles
infection or administration of measles-containing vaccines.23 Mantoux testing
is therefore unreliable for at least 4weeks after the administration of MMR
or MMRV. As measles infection may cause exacerbation of tuberculosis,
there is a theoretical concern that measles-containing vaccine may exacerbate
tuberculosis. Patients with tuberculosis should be under treatment when
vaccinated.
Children with egg allergy, even anaphylactic egg allergy, can be safely given
MMR or MMRV vaccine.2,34 Skin testing has been shown to be of no value
in the management of these cases.2 Although measles and mumps (but not
rubella or varicella) vaccine viruses are grown in chick embryo tissue cultures,

Measles 207

3.11 Measles

Children with a personal or close family history of seizures or convulsions


should be given MMR or MMRV vaccine, provided the parents understand
that there may be a febrile response 5 to 12days after vaccination.23 Advice
for reducing fever with paracetamol and other measures should be given.
Specialist advice should be sought rather than refusing to provide MMR or
MMRV vaccination.

it is now recognised that MMR (and MMRV) vaccine contains negligible


amounts of egg protein (see Variations from product information below).
MMR and MMRV vaccines can be administered to susceptible children who
have mild illnesses (eg. diarrhoea or upper respiratory infection), with or
without low-grade fever (<38.5C).

Adverse events
(If using MMRV vaccine, additional adverse events relating to the varicella
vaccine component are outlined in Chapter 3.24, Varicella.)
Malaise, fever and/or a rash may occur after MMR vaccination, most
commonly 7 to 10days (range 512days) after vaccination and lasting about
2 to 3days. High fever (>39.4C) occurs in approximately 5 to 15% (common
to very common), and rash occurs in approximately 5% (common) of MMR
vaccinees.1,23 The risk for febrile seizures is approximately 1 case per 3000
doses of MMR vaccine administered.23 Slightly higher rates of fever were
observed in clinical trials of MMRV vaccines, as compared with giving MMR
and monovalent varicella vaccine at the same time but at separate sites.24,25
A varicelliform rash may occur after vaccination with MMRV (see Chapter
3.24, Varicella Adverse events). The appearance of a rash after monovalent
varicella vaccine occurs in <5% (common) of vaccinees, and similar rates are
observed with the use of MMRV.35
Adverse events are much less common after the second dose of MMR and
MMRV than after the first dose.
Anaphylaxis following the administration of MMR is very rare (less than
1 in 1 million doses distributed).23 Although no cases of anaphylaxis were
reported in MMRV clinical trials, the incidence is likely to be similar to
that occurring with use of MMR. Anaphylaxis after vaccination is likely
due to gelatin or neomycin anaphylactic sensitivity, not egg allergies (see
Precautions above).
It is uncertain whether encephalopathy occurs after measles vaccination. If it
does, it is at least 1000 times less frequent than as a complication from natural
infection.1,23
Other rare adverse events attributed to MMR vaccine include transient
lymphadenopathy and arthralgia (see Chapter 3.19, Rubella). Parotitis has
been reported rarely.23
Thrombocytopenia (usually self-limiting) has been very rarely associated
with the rubella or measles component of MMR occurring in 3 to 5 per
100000 doses of MMR vaccine administered.2,23,32,33 This is considerably less
than after natural measles, mumps and rubella infections33 (see also Chapter
3.19, Rubella). Any association with MMRV vaccine is expected to be similar.

208 The Australian Immunisation Handbook 9th Edition

It is recommended that parents/vaccine recipients be advised about


possible symptoms, and given advice for reducing fever, including the use
of paracetamol for fever in the period 5 to 12days after vaccination. The use
of aspirin after MMRV vaccination is not recommended for 6weeks (see
Chapter 3.24, Varicella).
It had been hypothesised that the measles component of the MMR vaccine
may be causally linked with autism, autistic spectrum disorder and
inflammatory bowel disease.36 There has been no credible scientific evidence
to support this claim. Most proponents of the hypothesis have retracted
this claim37 and there is now good evidence to refute it38 (see Appendix 5,
Commonly asked questions about vaccination).

Transmissibility of MMR vaccine viruses


Measles, mumps and rubella vaccine viruses are not transmissible to contacts.23
It is, therefore, safe to vaccinate the healthy siblings of children with impaired
immunity and safe for children with impaired immunity to go to school with
children recently vaccinated with the MMR vaccine. If using MMRV, see Chapter
3.24, Varicella for information about varicella vaccine virus transmission.

The public health management of measles


(i) Definition of a person who is considered not susceptible to measles
A person is considered not susceptible to measles if he/she meets 1 of the
following criteria:

born before 1966 (unless serological evidence indicates otherwise),


documented evidence of immunity,
documented evidence of laboratory confirmed measles infection.
NB. These criteria have been revised since publication of the Guidelines for the
control of measles outbreaks in Australia in 2000.39

(ii) Vaccination of measles contacts


As vaccine-induced measles antibody develops more rapidly than that after
natural infection, MMR vaccine can be used to protect susceptible contacts.23 The
incubation period of the vaccine strain (4 to 6days) is shorter than the incubation
period of wild measles virus (10 to 14days). To be effective, the vaccine must
be administered within 72 hours of exposure. If there is doubt about a persons
immunity, vaccine should be given, since there are no ill effects from vaccinating
individuals who are already immune. It must be noted that antibody responses

Measles 209

3.11 Measles

born during or since 1966 with documented evidence of receiving 2 doses of a


measles-containing vaccine, with both doses of vaccine having been given at
12months of age and at least 4weeks apart. This applies unless serological
evidence indicates otherwise,

to the rubella and mumps components are too slow for effective use of vaccine
as prophylaxis after exposure to these infections. Alternatively, MMRV vaccine,
when available, could also be used in this setting if varicella vaccination
is indicated. However, there are no data on the use of MMRV vaccines in
individuals >12years of age.
Immunoglobulin is available for contacts for whom measles-containing vaccine
is contraindicated (see Use of immunoglobulin to prevent measles below), for
infants aged 69months, and for susceptible individuals who did not receive a
measles-containing vaccine within 72 hours of contact (see Table 3.11.1 below).
Isolation of susceptible close contacts by exclusion from school or the workplace
should occur until 14days after their last exposure39 unless they receive either
the MMR vaccine within 72 hours or immunoglobulin within 7days of their first
exposure. If they do not receive MMR vaccine or immunoglobulin within these
specified timeframes, they should be excluded.

(iii) Vaccination during an outbreak


During a confirmed measles outbreak, MMR vaccine may be given (on the
direction of public health authorities) to infants between 9 and 12months of age,
and even to those between 6 and 9months of age.39 In these cases, another dose of
MMR (or MMRV when available) should be given at 12months of age or 4weeks
after the first dose, whichever is later. This should be followed by the routine
administration of the next dose of measles-containing vaccine at 18months of
age. This is because maternal antibodies to measles are known to persist in many
infants until 11months of age and may interfere with active immunisation before
12months of age.1
Children between 12 and 18months of age who have received 1 dose of measlescontaining vaccine can be offered their second dose early (ie. at least 4weeks
after the first dose) if they are considered at risk of coming in contact with
measles39 (see Recommendations (i) above). If a child receives the second dose
early, he/she is considered to have completed the vaccination schedule and,
therefore, does not require another dose at 18months of age or beyond, provided
2 doses were given at 12months of age and at least 4weeks apart.
Any older children, adolescents or adults who are considered susceptible to measles
(see (i) above) during an outbreak should receive MMR (or MMRV if appropriate).

(iv) Use of immunoglobulin to prevent measles


Normal human immunoglobulin (NHIG) should be considered for contacts of
patients with confirmed or suspected measles39 (see Table 3.11.1). If NHIG is
administered by IM injection within 7days of exposure, it can prevent or modify
measles in non-immune individuals.
The dose of NHIG is 0.2mL/kg by deep IM injection for healthy children,
adolescents and adults (including pregnant women), and 0.5mL/kg by deep IM
injection for people with impaired immunity. The maximum dose is 15mL.

210 The Australian Immunisation Handbook 9th Edition

NHIG should be given to exposed individuals if contact was within the previous
7days in the following instances:
infants <6months of age where the infants mother is the measles case, or the
infant was born before 28weeks gestation,
infants 69months of age (see Vaccination during an outbreak above),
all those 9months of age in whom administration of MMR vaccine would
be contraindicated,
non-immune pregnant women,
those exposed to measles who have impaired immunity,
those who have never received a measles-containing vaccine, and who did
not receive a MMR or MMRV vaccine within 72 hours of contact.
Children with impaired immunity, where MMR is contraindicated, should be
given NHIG as soon as possible (within 7days) after exposure. Testing for measles
antibody does not assist with the decision to use immunoglobulin, since neither
previous vaccination nor demonstrated low-level serum antibody guarantees
immunity to measles in individuals with significantly impaired immunity.23,39
Testing for measles antibody may delay the appropriate use of NHIG. However,
testing may be of value in making a definitive diagnosis of measles.

Measles 211

3.11 Measles

Infants 69months of age who have direct contact with a person with measles
are at risk of developing complications from the disease, and should be
offered NHIG within 7days of contact.39 MMR vaccine should then be given
as close as possible to 12months of age, after an interval of at least 5months
following the administration of immunoglobulin (see Chapter 2.3, Groups with
special vaccination requirements, Table 2.3.5 Recommended intervals between either
immunoglobulins or blood products and MMR, MMRV or varicella vaccination). NHIG
is not usually given to babies <6months of age, who are protected by passive
maternal antibodies. However, if the mother of an infant <6months of age does
not have documented evidence of having received 2 doses of MMR, or is the
measles case, the infant should be given NHIG. Similarly, premature infants
(<28weeks gestation) have little or no acquisition of transplacental maternal
antibody, irrespective of the number of doses of MMR the mother has received,
and should also be offered NHIG (see Table 3.11.1).

Table 3.11.1: Management of significant measles exposure using vaccination or


normal human immunoglobulin (NHIG)
Age

Action

<6months

NHIG 0.2mL/kg* IM injection if mother has not received


2 documented doses of MMR, or the mother is the measles
case, or the infant was premature (<28weeks gestation)

69months

NHIG 0.2mL/kg IM injection*

10months

MMR or MMRV vaccine within 72 hours of exposure OR NHIG


0.2mL/kg IM injection* if 37days after exposure

* The dose of NHIG is 0.2mL/kg in immunocompetent individuals and 0.5mL/kg in those


with impaired immunity.
Immunoglobulin is not required if the person has received at least 1 measles-containing
vaccine at 12months of age or is assessed as being not susceptible (see (i) Definition of a
person who is considered not susceptible to measles above), unless the person has impaired
immunity.

Use in pregnancy
MMR vaccine is not recommended in pregnancy due to the theoretical risk of
transmission of the rubella component of the vaccine to a susceptible fetus.
Pregnancy should be avoided for 28days after vaccination30 (see Chapter 3.19,
Rubella and Chapter 2.3, Groups with special vaccination requirements, Table 2.3.1
Vaccinations in pregnancy).

Variations from product information


The product information recommends that women of child-bearing age should
be advised not to become pregnant for 3months after vaccination with MMR or
MMRV vaccines, whereas the NHMRC recommends 28days.30
The product information for Priorix states that people with a history of
anaphylactic or anaphylactoid reactions should not be vaccinated with Priorix,
but it is established that MMR vaccine can be given in this situation.23

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

212 The Australian Immunisation Handbook 9th Edition

3.12 Meningococcal disease


Bacteriology
Meningococcal disease is caused by the bacterium Neisseria meningitidis
(N.meningitidis or the meningococcus), a Gram-negative diplococcus. There are
13 known serogroups distinguished by differences in surface polysaccharides of
the outer membrane capsule. Meningococcal serogroups are designated by letters
of the alphabet. Globally, serogroups A, B, C, W135 and Y most commonly cause
disease. Meningococci can be further differentiated by differences in their outer
membrane proteins, which are referred to as serotypes and serosubtypes.1 More
recently, molecular typing has been used to further differentiate meningococci.
In Australia, serogroups B and C occur most frequently. There is no consistent
relationship between serogroup or type and virulence.2,3

Clinical features
Neisseria meningitidis can cause meningitis, septicaemia or a combination of the
two. Other localised infections, including pneumonia, arthritis and conjunctivitis,
may also occur but are uncommon. Septicaemia, with or without meningitis,
can be particularly severe. The overall mortality risk is high (about 10%) despite
appropriate antibiotic therapy.
N. meningitidis is carried and transmitted only by humans. There are no known
animal reservoirs. Asymptomatic respiratory tract carriage of meningococci is
present in about 10% of the population, and the prevalence may be higher when
groups of people occupy small areas of living space.2-8 Recent studies indicate
that there may be a number of factors which contribute to the increased risk
of contracting meningococcal disease, including exposure to smokers, recent
illness, living in crowded conditions and multiple intimate kissing partners.4-8
People with inherited disorders of phagocytosis associated with properdin
deficiency or absence of the terminal components of complement, as well as
individuals with functional or anatomical asplenia, have an increased risk of
meningococcal infection.1

Meningococcal disease 213

3.12 Meningococcal
disease

The disease is transmitted via respiratory droplets, and has an incubation


period of between 1 and 10days, but commonly 3 to 4days.4 The capacity of
meningococcal disease to have a fulminant and rapidly fatal course in previously
healthy (and usually young) individuals causes it to be greatly feared. Intensive
public health follow-up is required after each single case to conduct contact
tracing and to institute appropriate public health measures for contacts. As a
result of all these factors, this disease causes widespread community alarm and
generates significant media interest.9

Epidemiology
Meningococci cause both sporadic and epidemic disease throughout the world.
Serogroup A disease occurs predominantly in developing populations such
as those in Africa and Asia, while serogroup B is the major cause of sporadic
meningococcal disease in most developed countries. Serogroup C disease has a
more cyclic pattern of occurrence, and increased in incidence in the 1990s in some
developed countries such as Australia and the United Kingdom.4 Serogroup
C meningococci have also been occasionally associated with small clusters of
meningococcal disease cases in schools, universities and nightclubs in Australia
over the past 10years.10-15
As in other temperate climates, meningococcal disease cases occurring
in Australia tend to follow a seasonal trend, the majority of cases being
reported during late winter and early spring. The overall notification rate of
meningococcal disease to the National Notifiable Diseases Surveillance System
increased gradually from 1.8 per 100000 in 1991, to a peak of 3.5 per 100000 in
2001, but declined to 1.8 per 100000 in 2005.16 There are considerable differences
noted in the incidence of meningococcal disease between States and Territories,
with 5.4 cases per 100000 notified from the Northern Territory to 1.9 per 100000
reported for Queensland during 2005.16 These figures include meningococcal
disease cases which were diagnosed on clinical grounds alone, and those cases
that were confirmed by laboratory methods such as culture, serology or nucleic
acid testing of clinical material. In 2005, 369 cases were reported nationally, of
which 345 were laboratory confirmed.16,17 The majority of laboratory-confirmed
meningococcal cases were serogroup B (73%) and serogroup C (14.5%).17 There
has been a steady decline in serogroup C meningococcal disease among the
018years age group since the 2003 introduction of routine meningococcal C
vaccination and catch-up programs in this age group.18
Meningococcal disease can occur in any age group, but the majority of cases
occur in those <5years of age, with a secondary peak seen in the 1524years
age group. In Australia, meningococcal disease in the <5years age group is
due predominantly to infection with serogroup B meningococci; very few cases
of serogroup C meningococcal disease are now seen in this age group.17 In the
1519years age group, both serogroup B and C disease were seen before the
introduction of the meningococcal C conjugate vaccine in 2003.
In contrast to Australia, New Zealand has, over the past 14years, experienced
an epidemic of meningococcal disease which has been almost exclusively
associated with a particular strain of serogroup B (B:4:P1.7b,4).19,20 Meningococcal
disease rates in NZ rose from 1.5 cases per 100000 during 19891990 to 14.5
cases per 100000 in 2003.19 A meningococcal B outer membrane vesicle vaccine
(MeNZB), currently being used in New Zealand, is only effective against the
serotype and serosubtype of the New Zealand serogroup B strain and is not
available in Australia.20

214 The Australian Immunisation Handbook 9th Edition

Vaccines
There are 2 different types of meningococcal vaccine: the meningococcal C
conjugate vaccines (MenCCV) and the tetravalent meningococcal polysaccharide
vaccines (4vMenPV). The differences between these 2 types of vaccines lie in the
different way that each vaccine stimulates an immune response.
Other than the New Zealand specific vaccine, there is currently no vaccine
effective against serogroup B meningococcal disease although extensive research
is being undertaken in this area.

CONJUGATE VACCINES
Meningococcal C conjugate vaccines (MenCCV)
Meningitec Wyeth Australia Pty Ltd (meningococcal serogroup
CCRM197 conjugate vaccine). Each 0.5mL monodose vial contains 10g
N. meningitidis serogroup C oligosaccharide conjugated to approximately
15g of a non-toxic Corynebacterium diphtheriae CRM197 protein;
aluminium phosphate.
Menjugate Syringe CSL Biotherapies/Novartis Vaccines (meningococcal
serogroup CCRM197 conjugate vaccine). Lyophilised powder in a monodose
vial with a pre-filled diluent syringe. Each 0.5mL dose of reconstituted
vaccine contains 10g N. meningitidis serogroup C polysaccharide conjugated
to 12.525g of a non-toxic Corynebacterium diphtheriae CRM197 protein; 1.0mg
aluminium hydroxide. 5 or 10 monodose packs also available.
NeisVac-C Baxter Healthcare (meningococcal serogroup Ctetanus toxoid
protein conjugate vaccine). Each 0.5mL pre-filled syringe contains 10g
N.meningitidis serogroup C polysaccharide conjugated to 1020g of
tetanus toxoid protein; 0.5mg aluminium hydroxide. 10 or 20 monodose
packs available.

MenCCVs confer protection only against serogroup C disease. In these vaccines,


an oligo- or polysaccharide antigen is chemically linked (ie. conjugated) to a
carrier protein. Conjugation changes the nature of the antibody response from
a T cell-independent to a T cell-dependent response. The T cell help results in
improved antibody responses, especially in young children, greater functional
activity, and induction of immunological memory, probably resulting in longterm protection.

Meningococcal disease 215

3.12 Meningococcal
disease

In January 2003, the Australian Government commenced the National


Meningococcal C Vaccination Program which provided free MenCCV to all children
who turned 1 to 19years of age during 2003. MenCCV was also added to the
National Immunisation Program (NIP) schedule at 12months of age at that time.

In the United Kingdom, 98 to 100% of infants given 3 doses of MenCCV in


a 2, 3 and 4month schedule developed protective antibody titres after the
third dose,21,22 but evidence of waning immunity and vaccine failures led to a
booster dose being recommended for children vaccinated according to the 2,
3 and 4month schedule of MenCCV,23 which has now been altered to a 3-dose
schedule at 3, 4 and 12months of age.24 In Australia, although some children
have received MenCCV before 12months of age, this was according to a 2, 4,
6month schedule and there is no evidence of vaccine failure. NHMRC, therefore,
does not recommend recall for a booster dose in children previously vaccinated
before 12months of age (unless they have inherited defects of properdin or
complement, or functional or anatomical asplenia see Recommendations
below).
In children >12months of age, a single dose of MenCCV appears sufficient to
induce protective antibody responses. In children 1218months of age receiving
a single dose of MenCCV, 91 to 100% achieved serum bactericidal antibodies
(SBA) titres 1:8.25 In older children, seroconversion rates increase with age: 96%
of 3-year-olds, 98% of 45-year-olds, and 98% of 1417-year-olds achieved SBA
titres 1:8 after a single dose.25 In those children aged 1 year who have received
only a single dose of the meningococcal C conjugate vaccine, the duration of
immunity and the need for booster doses is not yet known. The Netherlands
routinely vaccinates with MenCCV at 14months of age, and data from 2002
onwards currently indicates that vaccination after the 1st birthday results in
longer protection than multiple doses in infancy.26

POLYSACCHARIDE VACCINES
Meningococcal polysaccharide vaccines (4vMenPV)
Mencevax ACWY GlaxoSmithKline (serogroup A, C, W135 and Y
meningococcal polysaccharide vaccine). Each 0.5mL monodose of the
reconstituted lyophilised vaccine contains 50g of each polysaccharide;
lactose. 10-dose vials in packs of 50 contain 0.25% phenol as a preservative.
Saline diluent for each vial.
Menomune Sanofi Pasteur Pty Ltd (serogroup A, C, W135 and Y
meningococcal polysaccharide vaccine). Each 0.5mL monodose
of the reconstituted lyophilised vaccine contains 50g of each
polysaccharide;lactose.
4vMenPVs provide protection against serogroups A, C, W135 and Y. The
vaccine induces antibodies in 10 to 14days in 90% of recipients >2years of age.
Immunity decreases markedly during the first 3years following a single dose of
vaccine, particularly in infants and young children. However, clinical protection
persists for at least 3years in school children and adults.

216 The Australian Immunisation Handbook 9th Edition

The duration of immunity is further complicated by the induction of


immunological hyporesponsiveness to the serogroup C component following
repeated vaccination with 4vMenPV, as revaccination results in a reduced
antibody response compared with the primary immunisation.27 This
phenomenon has been noted in both children and adults.28-31 The demonstration
of subsequent hyporesponsiveness has led to the concern that vaccinating lowrisk individuals may reduce the effectiveness of revaccination in a subsequent
high-risk situation, although this has not been clinically demonstrated. This
hyporesponsiveness can be overcome with MenCCV, although additional doses
of the conjugate vaccine may be required in young children.32,33 There is little
response to the serogroup C component of the 4vMenPV before 18months of age
and little response to serogroup A before 3months of age.34,35

Transport, storage and handling


Meningococcal C conjugate vaccines
Transport according to National Vaccine Storage Guidelines: Strive for 5.36 Storage of
all MenCCVs should be at +2C to +8C. Do not freeze. Protect from light.
The product information for NeisVac-C states that this vaccine can be stored at
+25C for a period of up to 9months but must not be returned to the refrigerator.

Meningococcal polysaccharide vaccines


Transport according to National Vaccine Storage Guidelines: Strive for 5.36 Store at
+2C to +8C. Do not freeze. Protect from light. Reconstituted vaccine should
be used immediately but may be stored in the refrigerator at +2C to +8C, and
must be discarded if not used within 8 hours.

Dosage and administration


Meningococcal C conjugate vaccines

Meningococcal polysaccharide vaccines


The 4vMenPV dose is 0.5mL, administered by SC injection. 4vMenPVs are
approved for use in Australia in children 2years of age, adolescents and adults.

Administration of MenCCV after administration of 4vMenPV


There are limited data available on the length of time that should lapse before
administration of MenCCV after giving 4vMenPV. The NHMRC recommends a
period of 6months before the conjugate vaccine is given.21,37

Meningococcal disease 217

3.12 Meningococcal
disease

The MenCCV dose is 0.5mL given by IM injection. Do not mix MenCCV with
other vaccines in the same syringe. Experience from the use of the conjugate Hib
vaccines suggests that the different brands of the MenCCVs are interchangeable.
MenCCVs may be administered simultaneously with other vaccines in the NIP
(see Variations from product information below). MenCCVs are registered for
use in Australia from 6weeks of age.

Administration of 4vMenPV after administration of MenCCV


On occasion, both MenCCV and 4vMenPV are recommended (eg. asplenia). If
MenCCV is given first, a period of 2weeks should lapse before 4vMenPV is given.

Recommendations
Meningococcal C conjugate vaccines
(i) Routine vaccination
It is recommended that a single dose be given to all children at the age of
12months.
Vaccination before 12months of age is not recommended, except in infants
with inherited defects of properdin or complement, or functional or anatomical
asplenia (see (ii) below). Infants, other than those described in the circumstances
below, who receive dose(s) of vaccine at <12months of age, should be given
a further dose at 12months of age or 4weeks after the last dose, whichever is
later. However, it is not necessary to recall older children who received 3 doses
of MenCCV before 12months of age, as there has been no evidence to date of
vaccine failure in infants vaccinated according to a 2, 4, 6month schedule (see
Vaccines above).

(ii) Vaccination of people at high risk for meningococcal disease


The vaccine is also recommended for:
close (household or household-like) contacts of meningococcal disease
cases due to serogroup C, >2months of age, who have not previously been
vaccinated (refer to The early clinical and public health management of
meningococcal disease below),
control of outbreaks caused by serogroup C (refer to The early clinical and
public health management of meningococcal disease below),
laboratory personnel who frequently handle N. meningitidis, who should also
receive 4vMenPV,
those >6weeks of age with inherited defects of properdin or complement,
or functional or anatomical asplenia. When MenCCV is given to these
individuals at <12months of age, in addition to a booster dose of MenCCV at
12months of age, a dose of 4vMenPV is recommended at 2years of age.
Under the circumstances as described above, a child under the recommended age
of 12months requiring vaccination should receive doses as follows:
Infants <6months of age require 2 doses of 0.5mL, given at least 8weeks
apart, followed by a booster dose at 12months of age.
Children 611months of age require 1 dose of 0.5mL, followed by a booster
dose at 12months of age or 8weeks after the first dose, whichever is later.

218 The Australian Immunisation Handbook 9th Edition

Meningococcal polysaccharide vaccines


Routine vaccination with 4vMenPV is not recommended. However, it is
recommended in the following situations:
people who intend travelling to parts of the world where epidemics of group
A, W135 or Y disease are frequent (a current list of those countries is available
from the World Health Organization at either http://www.who.int/ith or
http://www.who.int/disease-outbreak-news/),
close (household or household-like) contacts, 2years of age, of cases of
serogroup A, W135 or Y meningococcal disease,
control of outbreaks caused by serogroup A, W135 or Y. A Cochrane review
examined the use of polysaccharide vaccine for the prevention of serogroup
A meningococcal meningitis. The review assessed 8 randomised controlled
trials and the protective effect from the vaccine was consistent across all the
trials with a vaccine efficacy of around 95%,38
laboratory personnel who frequently handle N. meningitidis, who should also
receive MenCCV,
those 2years of age with inherited defects of properdin or complement, or
functional or anatomical asplenia, who should also receive MenCCV, and
pilgrims attending the annual Hajj (Saudi Arabian authorities require a valid
certificate of vaccination as a condition to enter the country).
A single revaccination with 4vMenPV is indicated for people at continued high
risk of infection (such as those living in epidemic areas, and those with impaired
immunity as defined above), particularly children first vaccinated before 4years
of age. As antibody levels decline rapidly over 2 to 3years, revaccination
should be given 3 to 5years later. Data regarding the benefit of subsequent
revaccinations with 4vMenPV are unavailable at this time.

Contraindications
Meningococcal C conjugate vaccines
The only absolute contraindications to MenCCV are:
anaphylaxis following a previous dose, or
Previous serogroup C disease is not a contraindication to administration of
MenCCV.

Meningococcal polysaccharide vaccines


The only absolute contraindications to 4vMenPV are:
anaphylaxis following a previous dose, or
anaphylaxis following any vaccine component.

Meningococcal disease 219

3.12 Meningococcal
disease

anaphylaxis following any component of the vaccine

Adverse events
Meningococcal C conjugate vaccines
Very common (>10%) adverse events caused by MenCCVs are pain, redness
and swelling at the site of injection, fever, irritability, anorexia and headaches.
There are some age-related differences in the type of adverse event following
vaccination, with systemic adverse events tending to decrease with increasing
age, and local adverse events tending to increase with increasing age. Headache
is more likely to be reported in the adolescent age group. Serious general adverse
events are rare.37

Meningococcal polysaccharide vaccines


Local reactions to 4vMenPV include erythema, induration, tenderness, pain and
local axillary lymphadenopathy. However, they are usually mild and infrequent.
Fever and chills occur in approximately 2% (common) of young children, and
may persist for 48 hours or longer, but significant general adverse events are rare.

The early clinical and public health


management of meningococcal disease
Prompt diagnosis and emergency treatment of cases of suspected meningococcal
disease are life-saving. If a diagnosis of meningococcal disease is suspected,
the patient should be immediately given parenteral (usually IM) penicillin and
transferred to hospital. The relevant Public Health Unit must be contacted as
soon as possible.4
Table 3.12.1: Early clinical management of suspected meningococcal disease
The patient should
receive immediate
benzylpenicillin
(usually IM).

Age <1 year

300mg benzylpenicillin

Age 19years

600mg benzylpenicillin

Age 10years

1200mg benzylpenicillin

The patient should be transferred to hospital urgently.


The relevant Public Health Unit should be notified promptly, so that
appropriate public health management can be initiated.

Guidance on the early clinical and public health management of


cases of invasive meningococcal disease has been developed by the
Communicable Diseases Network of Australia, and is available at
http://www.health.gov.au/pubhlth/cdi/pubs/pdf/mening_guide.pdf.4
Contrary to popular belief, a patient with meningococcal disease is not a good
transmitter of the disease. Rather, it is carrier(s) passing on the bacteria to other
susceptible individuals who may cause further cases of meningococcal disease.
Further cases may develop in household contacts in particular. The risk of
secondary cases is greatest in the first 7days, and may persist for manyweeks
after contact. The public health management of close contacts includes

220 The Australian Immunisation Handbook 9th Edition

information, clearance antibiotics and vaccination. Clearance antibiotics should


be offered to all identified close contacts regardless of previous vaccination
history. Clearance antibiotics are not recommended for healthcare workers unless
they were engaged in either mouth-to-mouth resuscitation or were not wearing a
mask while intubating a case.
Non-vaccinated close contacts of a proven vaccine-preventable strain of invasive
meningococcal disease should be advised in writing by their local Public Health
Unit to visit their usual healthcare provider at the next available opportunity to
receive the appropriate vaccine.4
Antibiotics that reduce or eliminate nasopharyngeal carriage of N. meningitidis
include ceftriaxone, ciprofloxacin and rifampicin.
Ceftriaxone is administered as a single IM dose of 250mg for adults and
125mg for children <12years of age. Although it is considerably more
expensive, ceftriaxone has a number of advantages over rifampicin: it is
more likely to eradicate pharyngeal carriage, it eliminates problems with
compliance and it is the preferred chemoprophylaxis for pregnant women.
Ciprofloxacin in a single oral dose of 500mg is effective and safe, but it
should not be given to children <12years of age, or to pregnant women.
Rifampicin is given to children and adults in an oral dose of 10mg/kg
(maximum dose of 600mg) twice daily for 2days. The recommended dose
for infants <1month of age is 5mg/kg twice daily for 2days. Pharyngeal
carriage will be eliminated in 75 to 90% of recipients unless the strain is
resistant to rifampicin. The side effects of rifampicin should be explained,
including orange-red discolouration of contact lenses, urine and tears,
possible interference with the contraceptive pill, and interference with the
metabolism of many other drugs including warfarin, phenobarbitone and
phenytoin. Rifampicin is not recommended for use in pregnant women.

Use in pregnancy
Meningococcal C conjugate vaccines
Although no clinical study data are available on the use of MenCCV in pregnant
women, it is unlikely that it would have any deleterious effect on the pregnancy.
Routine pregnancy testing before vaccination is not justified.

Meningococcal disease 221

3.12 Meningococcal
disease

A potential outbreak of meningococcal disease in an institutional or community


setting is a public health emergency needing a rapid response from clinicians
and public health practitioners. The decision to control an outbreak with a
vaccination program should be made by the appropriate Public Health Unit,
following the Guidelines for the early clinical and public health management of
meningococcal disease in Australia.4 If vaccination is indicated and the organism
responsible is serogroup C, MenCCV should be used in preference to 4vMenPV.

Meningococcal polysaccharide vaccines


Studies of vaccination with meningococcal and other polysaccharide vaccines
during pregnancy have not documented adverse events in either pregnant
women or newborns.1,39,40 The number of pregnant vaccinees who received
4vMenPV as reported in the literature is small. A North American study of
109 women who received the 4vMenPV vaccine between 30 and 38weeks
gestation reported no birth defects.39 A further study of 34 pregnant women in
the US who received 4vMenPV during their second and third trimester revealed
no teratogenicity and the infants were assessed for 2years after birth. No
developmental abnormalities were detected.40

Variations from product information


Meningococcal C conjugate vaccines
The product information for meningococcal C conjugate vaccines state
that, under the age of 12months, either 2 (NeisVac-C) or 3 (Meningitec and
Menjugate) doses of vaccine are required. The NHMRC recommends that
meningococcal C vaccination is not required before 12months of age (unless
specifically indicated).
The NeisVac-C product information states that the vaccine should not be
administered with pneumococcal conjugate vaccine, hepatitis B vaccine
and PRP-OMP Haemophilus influenzae type b vaccine unless medically
important. However, the NHMRC states that the vaccine may be administered
simultaneously with other vaccines in the NIP. There have been recent
publications citing the coadministration of MenCCV with other combination
vaccines and it was found to be immunogenic and safe.41,42
The product information for all 3 conjugate vaccines states that there are no data
on the use of MenCCVs in lactating women, whereas the NHMRC does not
consider breastfeeding in a healthy woman a reason for not vaccinating.
The Meningitec product information states that an allergic reaction following
a previous dose is a contraindication to further doses, whereas the NHMRC
states that only an anaphylactic adverse event following a previous dose is a
contraindication.

Meningococcal polysaccharide vaccines


The NHMRC recommends revaccination with 4vMenPV within 3 to 5years
of a previous dose in the situations detailed in Recommendations above.
The Mencevax ACWY product information states within 2 to 3years, and
the Menomune product information gives no recommended interval before
revaccination.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

222 The Australian Immunisation Handbook 9th Edition

3.13 Mumps

Mumps is a paramyxovirus, genus Rubulavirus, with a single-stranded RNA


genome. It is rapidly inactivated by heat, formalin, ether, chloroform and
ultraviolet light.1

Clinical features
Mumps is an acute viral illness with an incubation period of 14 to 25days.2
Asymptomatic infection occurs in one-third of cases.3 Symptomatic disease
ranges from mild upper respiratory symptoms to widespread systemic
involvement.3 A high proportion of mumps infections involve non-specific
symptoms including fever, headache, malaise, myalgia and anorexia.4 The
characteristic bilateral, or occasionally unilateral, parotid swelling occurs in 60
to 70% of clinical cases.4 Benign meningeal signs appear in up to 15% of cases,
but permanent sequelae are rare. Nerve deafness is one of the most serious of the
rare complications (1 in 500 hospitalised cases). Orchitis (usually unilateral) has
been reported in up to 20% of clinical mumps cases in post-pubertal males, but
subsequent sterility is rare. Symptomatic involvement of other glands and organs
has been observed less frequently (pancreatitis, oophoritis, hepatitis, myocarditis,
thyroiditis, mastitis).1,4 Patients may be infectious from 6days before parotid
swelling to 9days after.2 Mumps encephalitis has been reported to range as high
as 1 in 200 cases, with a case-fatality rate of around 1.0%.
Mumps infection during the first trimester of pregnancy may result in
spontaneous abortion.3,4 Maternal infection is not associated with an increased
risk of congenital malformation.3,4

Epidemiology
Mumps is reported worldwide, and is a human disease. Transmission is by
the respiratory route in the form of air-borne droplets or by direct contact with
saliva or possibly urine.2 Before universal vaccination, mumps was primarily
a disease of childhood, the peak incidence being in the 59 year age group.
However, since 2000, peak rates have been reported in older adolescents and
young adults, especially the 2024 year age group.5-7 Between 2001 and 2005, the
average notification rate for mumps in Australia was 0.6 per 100000.8 There have
been recent outbreaks of mumps in the USA, and also in the United Kingdom,
where the peak rates of disease have been in the 1824 year age group.9,10
Approximately 2000 cases were reported in the USA in a 2006 outbreak, parotitis
being reported in 66% of clinical cases.11 Mumps attack rates in outbreaks are
lowest in individuals who have received 2 doses of mumps-containing vaccines,
as this provides optimal long-term protection.10,11 In Australia, over the 10-year

Mumps 223

3.13 Mumps

Virology

period from 1996 to 2005, mumps has been reported as the underlying cause of
death in 4 subjects, all adults aged over 80years.5-7

Vaccines
Monovalent mumps vaccine is no longer available in Australia. Mumps
vaccination is provided using either MMR vaccine or MMRV vaccine when
available.
Clinical trials of mumps vaccine indicate 95% seroconversion after a single dose
of MMR.4 However, outbreak investigations and post-marketing studies have
reported 1-dose vaccine effectiveness to be between 60 and 90%.10 Protection
is greater in 2-dose vaccine recipients, who have seroconversion rates of up to
100%.4,10,12
Priorix (MMR) GlaxoSmithKline (live attenuated measles virus (Schwarz
strain), RIT 4385 strain of mumps virus (derived from the Jeryl Lynn strain)
and the Wistar RA 27/3 rubella virus strain). Each 0.5mL monodose of the
reconstituted, lyophilised vaccine contains not less than 103.0 CCID50 (cell
culture infectious dose 50%) of the Schwarz measles, not less than 103.7
CCID50 of the RIT 4385 mumps and not less than 103.0 CCID50 of the Wistar
RA 27/3 rubella virus strains; lactose; neomycin; amino acids; sorbitol and
mannitol as stabilisers.

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.13 Store at
+2C to +8C. Protect from light. Do not freeze. Reconstituted vaccine should be
used immediately, but can be stored at +2C to +8C for up to 8 hours before use.

Dosage and administration


For both children and adults, the dose of MMR is 0.5mL, administered by either
SC or IM injection.
MMR can be given at the same time as other vaccines (including DTPa, hepatitis
B, MenCCV and varicella), using separate syringes and injection sites. If MMR is
not given simultaneously with other live viral parenteral vaccines (eg. varicella
vaccine), they should be given at least 4weeks apart (see Precautions below).

Recommendations
All children should receive 2 doses of MMR vaccine (or MMRV vaccine, when
available, if 12years of age). Routine administration of MMR (or MMRV) is
now recommended at 12months and 18months of age in order to maximise
protection at an early age. The minimum interval between doses is 4weeks.
In older individuals, who have received only 1 dose of mumps-containing
vaccine, a second dose can be given, as MMR, at any age.

224 The Australian Immunisation Handbook 9th Edition

For further information on the recommendations for MMR (and MMRV when
available) see Chapter 3.11, Measles and Chapter 3.24, Varicella.

See information on MMR and MMRV vaccines in Chapter 3.11, Measles and
Chapter 3.24, Varicella.

Precautions
If MMR is not given simultaneously with other live viral parenteral vaccines (eg.
varicella vaccine), they should be given at least 4weeks apart.
See further information on MMR and MMRV vaccines in Chapter 3.11, Measles
and Chapter 3.24, Varicella.

Adverse events
In Australia, vaccine-associated aseptic meningitis is not considered a problem.
Post-licensure surveillance of mumps vaccine recipients in Germany, over
a 2-year period, found no increase in cases of aseptic meningitis. However,
other estimates in countries using mumps vaccines with different vaccine
virus strains suggest 1 case occurs per 8000001 million doses.4,14 Vaccineassociated meningoencephalitis is invariably mild or asymptomatic and resolves
spontaneously. When mumps virus is isolated from the cerebrospinal fluid in
such cases, laboratory tests can be undertaken to distinguish between wild and
vaccine strains. The assistance of State virology laboratories should be sought in
such cases.
Re-vaccination with mumps-containing vaccines is not associated with an
increased incidence of adverse events.
For further information on the adverse events associated with MMR and MMRV,
see Chapter 3.11, Measles and Chapter 3.24, Varicella.

Use of immunoglobulin to prevent mumps


Normal human immunoglobulin (NHIG) has not been shown to be of value
in post-exposure prophylaxis for mumps.1,15 Live mumps-containing vaccine
does not provide protection if given after an individual has been exposed to
mumps.1,15 However, if the exposure did not result in infection, the vaccine
would induce protection against subsequent infection.

Use in pregnancy
MMR vaccine is not recommended in pregnancy due to the theoretical risk
of transmission of the rubella component of the vaccine to a susceptible fetus
(see Chapter 3.19, Rubella). Pregnancy should be avoided for 28days after
vaccination.16 Data on the use of MMRV vaccines in individuals >12years of age
are not available.

Mumps 225

3.13 Mumps

Contraindications

Variations from product information


See information on MMR vaccines in Chapter 3.11, Measles.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

226 The Australian Immunisation Handbook 9th Edition

3.14 Pertussis
Bacteriology
Pertussis (whooping cough) is caused by Bordetella pertussis, a fastidious, Gramnegative, pleomorphic bacillus. There are other organisms (such as Bordetella
parapertussis, Mycoplasma pneumoniae and Chlamydia pneumoniae) which can cause
a pertussis-like syndrome.1 Identification of pertussis is limited by patient and
physician awareness and the limited sensitivity of diagnostic tests; it is generally
believed to be significantly under-diagnosed.

Clinical features

Death due to pertussis is rare in people >10years of age. However, the case
fatality rate in unvaccinated infants <6months of age is estimated to be
0.8%.5,6 The most common cause of death in pertussis infection is pertussis
pneumonia, sometimes complicated by seizures and hypoxic encephalopathy.2
Both hospitalisations and deaths are likely to be under-estimated.7 In Australia
between 1993 and 2005, there were 18 deaths attributed to pertussis, all but 2 in
infants <12months of age.8-11

Epidemiology
Epidemics occur every 3 to 4years. In unvaccinated populations, these outbreaks
can be very large. In vaccinated populations, outbreaks are smaller, with greatly
reduced mortality and morbidity, and may continue to occur every 3 to 4years or
be more widely spaced. Maternal antibody does not provide reliable protection
against pertussis, and the maximal risk of infection and severe morbidity is
before infants are old enough to have received at least 2 vaccine doses.7 In
recentyears, among highly immunised communities, many cases of pertussis
in adults and adolescents, due to waning immunity, have been recognised due
to the increased availability of serological testing.6,12 These individuals are a
significant reservoir of infection. From 1993 to 2005, 4 epidemics of pertussis

Pertussis 227

3.14 Pertussis

Pertussis is an epidemic respiratory infection with an incubation period of


7 to 20days. In unvaccinated individuals, B. pertussis is highly infectious,
spreading by respiratory droplets to 80% of susceptible household contacts.2 The
characteristic paroxysmal cough with inspiratory whoop seen in unvaccinated
children is less common in older children and adults who have varying degrees
of immunity acquired from vaccination or infection. It has been estimated that
B. pertussis accounts for up to 7% of cough illness per year in adults and, each
year, more than 25% of adults experience a coughing illness of at least 5days
duration.2,3 Even in adults, pertussis can be associated with significant morbidity,
with cough persisting for up to 3months, and other significant symptoms, such
as sleep disturbance or, rarely, rib fracture.4

occurred in Australia. A total of more than 84000 cases was reported in this time,
an annual incidence of 22.8 to 57.4 cases per 100000 population.13
Introduction of a fifth dose of diphtheria, tetanus and pertussis vaccine (DTP) for
45-year-old children in August 1994 was followed by a pattern of decrease in
notifications consistent with a vaccine effect, occurring first among children aged
5 and 6years, followed by those in the 79-year age group.14,15 Subsequently, the
average age of those notified with pertussis has continued to increase.
In 20042005, more than 70% of pertussis notifications were in individuals
>15years of age13,16 compared with 40 to 50% in the early 1990s. This supports
the need for booster doses in those >10years of age, both to reduce individual
morbidity, and to reduce transmission to those most at risk (infants <6months
of age).17 Vaccination of adolescents, who have a high risk of pertussis infection,
and adults in contact with very young infants, is expected to result in the greatest
health benefits. A single booster dose using an adolescent/adult formulation
acellular pertussis-containing vaccine (dTpa) has been included in the National
Immunisation Program (NIP) for 1517-year-olds since January 2004.
Figure 3.14.1: Pertussis notifications by year of onset, Australia 19932005. The
percentage of notifications in those aged 15years is shown (- -)

228 The Australian Immunisation Handbook 9th Edition

Vaccines
Acellular pertussis-containing vaccines have been used for both primary and
booster vaccination of children in Australia since 1999. There are a number
of acellular pertussis vaccines, which contain 3 or more purified components
of B. pertussis. In the 3 component vaccines, these are pertussis toxin (PT),
filamentous haemagglutinin (FHA) and pertactin (PRN). In the 5 component
vaccines, fimbrial antigens or agglutinogens are also included. Acellular pertussis
vaccines with 3 or more antigens have similar efficacy to good quality wholecell vaccines18 and are immunogenic and safe. Although serious adverse events
such as hypotonic-hyporesponsive episodes can still occur, they are much
less common than with whole-cell vaccines.18 Vaccines containing DTPa are
also available in various combinations with inactivated polio, hepatitis B and
Haemophilus influenzae type b vaccines.

The adolescent/adult formulation dTpa vaccines are immunogenic, safe and well
tolerated in adults.19-21
Formulations for children aged <8years
Infanrix hexa GlaxoSmithKline (DTPa-hepB-IPV-Hib; diphtheria-tetanusacellular pertussis-hepatitis B-inactivated poliomyelitis vaccine-Haemophilus
influenzae type b (Hib)). The vaccine consists of both a 0.5mL pre-filled syringe
containing 30 IU diphtheria toxoid, 40 IU tetanus toxoid, 25g pertussis
toxoid (PT), 25g filamentous haemagglutinin (FHA), 8g pertactin (PRN),
10g recombinant HBsAg, 40 D-antigen units inactivated polioviruses type
1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32 D-antigen units type 3
(Saukett) adsorbed onto aluminium hydroxide/phosphate; phenoxyethanol
as preservative; traces of formaldehyde, polymyxin and neomycin and a vial
containing a lyophilised pellet of 10g purified Hib capsular polysaccharide
(PRP) conjugated to 2040g tetanus toxoid. The vaccine must be reconstituted
by adding the entire contents of the syringe to the vial and shaking until the
pellet is completely dissolved. May also contain yeast proteins.
Infanrix-IPV GlaxoSmithKline (DTPa-IPV; diphtheria-tetanus-acellular
pertussis-inactivated poliomyelitis vaccine). Each 0.5mL pre-filled syringe
contains 30 IU diphtheria toxoid, 40 IU tetanus toxoid, 25g PT, 25g FHA,
8g PRN, 40 D-antigen units inactivated polioviruses type 1 (Mahoney),
8 D-antigen units type 2 (MEF-1) and 32 D-antigen units type 3 (Saukett)
adsorbed onto aluminium hydroxide; phenoxyethanol as preservative;
traces of formaldehyde, polymyxin and neomycin.

Pertussis 229

3.14 Pertussis

The acronym DTPa, using capital letters, signifies child formulations of


diphtheria, tetanus and acellular pertussis-containing vaccines. The acronym
dTpa is used for adolescent/adult formulations which contain substantially
lesser amounts of diphtheria toxoid and pertussis antigens (see formulations).

Infanrix Penta GlaxoSmithKline (DTPa-hepB-IPV; diphtheria-tetanusacellular pertussis-hepatitis B-inactivated poliomyelitis vaccine). Each
0.5mL pre-filled syringe contains 30 IU diphtheria toxoid, 40 IU tetanus
toxoid, 25g PT, 25g FHA, 8g PRN, 10g recombinant HBsAg, 40
D-antigen units inactivated polioviruses type 1 (Mahoney), 8 D-antigen
units type 2 (MEF-1) and 32 D-antigen units type 3 (Saukett) adsorbed onto
aluminium hydroxide/phosphate; phenoxyethanol as preservative; traces of
formaldehyde, polymyxin and neomycin. May also contain yeast proteins.
Formulations for people aged 8years
Combination vaccines
Adacel Sanofi Pasteur Pty Ltd (dTpa; diphtheria-tetanus-acellular
pertussis). Each 0.5mL monodose vial contains 2 IU diphtheria toxoid, 20
IU tetanus toxoid, 2.5g PT, 5g FHA, 3g PRN, 5g pertussis fimbriae
(FIM) 2+3; 1.5mg aluminium phosphate; phenoxyethanol as preservative;
traces of formaldehyde.
Adacel Polio Sanofi Pasteur Pty Ltd (dTpa; diphtheria-tetanus-acellular
pertussis-inactivated poliomyelitis vaccine). Each 0.5mL monodose vial
contains 2 IU diphtheria toxoid, 20 IU tetanus toxoid, 2.5g PT, 5g
FHA, 3g PRN, 5g FIM 2+3, 40 D-antigen units inactivated polioviruses
type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32 D-antigen
units type 3 (Saukett); 1.5mg aluminium phosphate; phenoxyethanol
as preservative; traces of formaldehyde, polymyxin, neomycin and
streptomycin.
Boostrix GlaxoSmithKline (dTpa; diphtheria-tetanus-acellular pertussis).
Each 0.5mL monodose vial or pre-filled syringe contains 2 IU diphtheria
toxoid, 20 IU tetanus toxoid, 8g PT, 8g FHA, 2.5g PRN, adsorbed
onto 0.5mg aluminium hydroxide/phosphate; 2.5mg phenoxyethanol as
preservative. May contain traces of formaldehyde.
Boostrix-IPV GlaxoSmithKline (dTpa-IPV; diphtheria-tetanus-acellular
pertussis-inactivated poliomyelitis vaccine). Each 0.5mL pre-filled syringe
contains 2 IU diphtheria toxoid, 20 IU tetanus toxoid, 8g PT, 8g FHA,
2.5g PRN, 40 D-antigen units inactivated polioviruses type 1 (Mahoney),
8 D-antigen units type 2 (MEF-1) and 32 D-antigen units type 3 (Saukett)
adsorbed onto aluminium hydroxide/phosphate; traces of formaldehyde,
polymyxin and neomycin.

230 The Australian Immunisation Handbook 9th Edition

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.22 Store at
+2C to +8C. Do not freeze. Protect from light.

Dosage and administration


The dose of DTPa-combinations, dTpa and dTpa-IPV is 0.5mL by IM injection.
Do not mix DTPa-containing vaccines with any other vaccine in the same
syringe, unless specifically registered for use in this way.

Recommendations
(i) Vaccination of children and adolescents

For the booster dose of DTPa given at 4years of age, all brands of DTPacontaining vaccines are considered interchangeable. In view of the prolonged
immunity now known to result from a primary course of DTPa at 2, 4 and
6months of age,23 the 18-month dose was omitted in 2003. It is expected that
postponing receipt of a fourth dose of DTPa until 4years of age will reduce the
proportion of children experiencing extensive local reactions, which occurred in
2% of children following a fourth dose at 18months of age.24,25
A second booster, between 12 and 17years of age, using the adolescent/adult
formulation dTpa, is essential for maintaining immunity to pertussis through the
adolescent period and into early adulthood. By the age of 17years, young adults
should have received 5 doses of a pertussis-containing vaccine. Most adolescents
would have either had at least 3 previous doses of a pertussis-containing vaccine
or been exposed to the pertussis bacterium. Therefore, if documentation of
previous vaccinations is not readily available, it can be safely assumed that a
dose of dTpa at 1217years is indeed a booster dose.
For details on the management of children who have missed doses in the NIP
schedule, see Section 1.3.5, Catch-up.

(ii) Vaccination of adults


dTpa vaccines are recommended in Australia for booster vaccination of
individuals 8years of age who have previously had a primary course of

Pertussis 231

3.14 Pertussis

Acellular pertussis antigens are given in combination with diphtheria and


tetanus toxoids (DTPa) in a primary course of vaccination at 2, 4 and 6months
of age. In view of the high morbidity and occasional mortality associated with
pertussis under the age of 6months, receipt of the first dose of vaccine as soon
as possible after 2months of age should be strongly emphasised. If the primary
course is interrupted, it should be resumed but not repeated; catch-up doses may
be given as little as 4weeks apart. The same formulation of vaccine should be
used for each of the 3 doses. If it is not known which brand was used, vaccination
should be provided using any available brand.

diphtheria-tetanus-pertussis vaccine. dTpa vaccines have a lower content of


diphtheria and pertussis antigens than DTPa formulations for young children.
Primary vaccination
If a 3-dose primary course of diphtheria/tetanus toxoids is given to an
adolescent/adult without a previous history of having received pertussiscontaining vaccine, the preferred option is that dTpa replace the first dose of dT, to
provide pertussis immunity as early as possible,26 with subsequent doses as dT. In
the event that dT is not available, dTpa can be used for all primary doses, but this
is not routinely recommended as there are no data on the safety, immunogenicity
or efficacy of dTpa for primary vaccination. For detailed recommendations
regarding a primary dT course in adults, see Chapter 3.21, Tetanus.
Duration of protection and spacing of booster doses
A single booster dose of dTpa is recommended for the following groups
provided that no documented dTpa booster dose has been previously received:
Adults planning a pregnancy, or for both parents as soon as possible
after delivery of an infant (preferably before hospital discharge), unless
contraindicated.17 Other adult household members, grandparents and carers
of young children should also be vaccinated. This recommendation is based
on evidence from several studies of infant pertussis cases, which indicated
that family members, particularly parents, were identified as the source
of infection in more than 50% of cases and were the presumed source in a
higher proportion.27-29
Adults working with young children. Vaccination is especially recommended
for childcare workers (see Chapter 2.3, Groups with special vaccination
requirements, Table 2.3.6 Recommended vaccinations for those at risk of
occupationally acquired vaccine-preventable diseases).30,31
All healthcare workers (see also Chapter 2.3, Groups with special vaccination
requirements, Table 2.3.6 Recommended vaccinations for those at risk of
occupationally acquired vaccine-preventable diseases). Several case reports
have documented nosocomial infection in young infants acquired from
healthcare workers.30,31
Any adult expressing an interest in receiving a booster dose of dT vaccine
should be encouraged to do so with dTpa vaccine. At the age routinely
recommended for tetanus and diphtheria booster (50years), dTpa produces
immune responses to tetanus and diphtheria antigens equivalent to dT
vaccine, and would also provide protection against pertussis.32
Data on the duration of immunity to pertussis following a single booster
dose of dTpa are limited in adults and adolescents.20,33 Although subsequent
doses of dTpa may prove beneficial, especially in high-risk groups such as
healthcare workers, such boosters are unlikely to be required before 10years and
recommendations must await further data.

232 The Australian Immunisation Handbook 9th Edition

Minimum interval between dTpa and other tetanus/


diphtheria-containing vaccines
A single dose of dTpa can be administered at any time after a dose of a vaccine
containing tetanus and diphtheria toxoids.
Recent studies from Canada have shown that a single dose of dTpa can be safely
administered as soon as 18months after a previous dose of a vaccine containing
tetanus or diphtheria toxoids.34-36 Where a tetanus- or diphtheria-containing
vaccine has been given even less than 18months previously, the benefits of
protection against pertussis are likely to outweigh the risk of an adverse event,37
and justify vaccination with dTpa or dTpa-IPV.

Special considerations
Previous pertussis infection

Pre-existing neurological disease and pertussis vaccination


Infants and children known to have active or progressive neurological disease
can be safely vaccinated with DTPa-containing vaccines. A large Canadian study
found no evidence of encephalopathy following acellular pertussis vaccines.38 For
infants and children with stable neurological disease (including cerebral palsy),
or a family history of idiopathic epilepsy or other familial neurological disorder,
the risk of adverse events following DTPa-containing vaccines are essentially the
same as for other infants of the same age.

Contraindications
The only true contraindications to acellular pertussis vaccines are:
anaphylaxis following a previous dose of an acellular pertussis vaccine, or
anaphylaxis following any vaccine component.

Precautions
Children who have a hypotonic-hyporesponsive episode (defined in Adverse
events below) following DTPa-containing vaccines should receive further
doses as scheduled in the National Immunisation Program. Supervision may be
required under some circumstances and specialist advice can be obtained from a
clinic specialising in the assessment and management of putative adverse events

Pertussis 233

3.14 Pertussis

Vaccination with pertussis vaccine in children, adolescents or adults who have


had laboratory-confirmed pertussis infection is safe but will not confer any
additional protection. If there is any uncertainty about a previous diagnosis
of pertussis, then vaccinate. In particular, incompletely vaccinated infants
<6months of age who develop pertussis may not mount an adequate immune
response following infection and should receive all routinely scheduled vaccines,
including pertussis.

following vaccination (see Appendix 1, Contact details for Australian, State and
Territory Government health authorities and communicable disease control).
A history of extensive limb swelling after a booster dose of DTPa is not a
contraindication to adolescent/adult formulation dTpa at 1217years of age
(or older).24 Parents of children about to receive the booster dose of a DTPacontaining vaccine (at 4years of age) should be informed of the small but welldefined risk of this adverse event which, even when extensive, is usually not
associated with significant pain or limitation of movement.

Adverse events
Significant adverse events following pertussis vaccination should be reported as
set out in Section 1.5.2, Adverse events following immunisation.
Acellular pertussis vaccines are associated with a much lower incidence of fever
(approximately 20%, very common) and local adverse events (approximately
10%, common) than whole-cell pertussis vaccines (approximately 45% and 40%,
respectively) which are no longer used in Australia).18,39
Following the introduction of DTPa in Australia, there was an increase
in the incidence of extensive local adverse events in children receiving
booster doses at 18months and 4years of age.40
Extensive limb swelling, defined by swelling and/or redness involving at
least half the circumference of the limb, and the joints both above and below
the injection site, was common (occurring in 2% of vaccinees) following a
booster dose of DTPa given at 18months of age; this was 1 reason for ceasing
this booster dose in 2003. Although it is still too early to assess the effect
that removing the 18-month booster dose has had on the incidence of local
adverse events following the booster dose at 4years of age, recent anecdotal
reports of much less extensive swelling are encouraging.41
Such reactions commence within 48 hours of vaccination, last for 1 to 7days
and resolve completely without sequelae.25 The pathogenesis of extensive
limb swelling is poorly understood. In an analysis of fourth and fifth dose
follow-up studies that examined 12 different DTPa vaccines, 2% (common)
of 1015 children who received consecutive doses of the same DTPa vaccine
reported entire thigh swelling, which resolved completely.25
Children who experience a febrile convulsion after a dose of DTPa-containing
vaccines are at increased risk (albeit low) of further febrile convulsion
following a subsequent dose of DTPa-containing vaccines. These risks can be
minimised by appropriate measures to prevent fever, so vaccination is still
recommended.
Febrile convulsions are very infrequently reported following DTPacontaining vaccines. The risk is even lower in infants who complete their
primary course at 6months of age, as febrile convulsions are uncommon
under 6months of age.

234 The Australian Immunisation Handbook 9th Edition

Hypotonic-hyporesponsive episodes (HHE), defined by an episode of


pallor, limpness and unresponsiveness 1 to 48 hours after vaccination, often
preceded by irritability and fever, occur rarely following DTPa. Shallow
respiration and cyanosis may also occur in an HHE. The whole episode lasts
from a few minutes to 36 hours. In Australia during 2005, 1.33 cases of HHE
were reported per 100000 doses of DTPa or DTPa-hepB vaccines given.41
Follow-up of children with HHE shows no long-term neurological or other
sequelae and they can receive further doses of DTPa-containing vaccines.42
Pertussis vaccine does not cause infantile spasms or epilepsy.
Sudden infant death syndrome (SIDS) is not associated with either DTPa or
any pertussis-containing vaccine.43 Some studies suggest a decreased risk of
SIDS in children who have been vaccinated (see Appendix 5, Commonly asked
questions about vaccination).

(i) Management of cases


The clinical case definition of pertussis is either (i) an acute cough lasting
14days with at least one of post-tussive vomiting, apnoea or whoop, or (ii)
a cough of any duration in a person epidemiologically linked to a laboratoryconfirmed case. The diagnosis can be definitively confirmed by either culture
or PCR of a per-nasal swab or nasopharyngeal aspirate. The serological tests
available in most areas of Australia are based on detection of IgA antibodies
to B. pertussis antigens and are insensitive, so that false negative results are a
problem, especially if performed on only one occasion.44 In those who have not
received a pertussis-containing vaccine within the previous 5years, detection
of IgA to pertussis antigens is highly specific in the presence of appropriate
symptoms. If pertussis is suspected in someone who has received a pertussiscontaining vaccine within 5years, PCR is the diagnostic method of choice, but
has progressively decreasing sensitivity with increased duration of symptoms.
In a research setting, an IgG PT level of at least 125 EU/mL has been shown to
be indicative of a recent or active pertussis infection; however, this assay is not
available in routine pathology laboratories.45,46
A detailed history is required when a case of pertussis is suspected, including
date of onset, vaccination status and details of household contacts. To reduce the
risk of transmission, cases should be commenced on antibiotic therapy on clinical
suspicion, but only if commenced within 21days of the onset of coryza. There
is no evidence of any reduction in pertussis transmission following antibiotic
treatment if the case has had symptoms for more than 21days. Appropriate
macrolide antibiotics for treatment of pertussis are azithromycin, clarithromycin
and erythromycin. An alternative for those unable to take macrolides is
trimethoprim-sulfmethoxazole. Table 3.14.1 shows the dose regimens for each of
these antibiotics. (See also Variations from product information below.)

Pertussis 235

3.14 Pertussis

The public health management of pertussis

Table 3.14.1: Recommended antimicrobial therapy and chemoprophylaxis


regimens for pertussis in infants, children and adults47-54
Age group

Azithromycin

Clarithromycin

Erythromycin

TMP-SMX*

<1month

10mg/kg single
dose for 5days

Not
recommended

If azithromycin
is unavailable;

Not
recommended
in infants
<2months of age
unless macrolides
cannot be used

7days old:
10mg/kg/
dose 12-hourly
for 7days;
828days old:
10mg/kg/
dose 8-hourly
for 7days
15months

10mg/kg single
dose for 5days

7.5mg/kg/
dose twice daily
for 7days

10mg/kg/
dose 6-hourly
for 7days

2months of
age; TMP: 4mg/
kg twice daily,
SMX: 20mg/
kg twice daily
for 7days

Infants
(6months)
and children

10mg/kg single
dose on day
1, then 5mg/
kg single dose
fordays 25

7.5mg/kg/
dose (up to a
maximum dose
of 500mg) twice
daily for 7days

(maximum
250mg/day)

(maximum
1g/day)

10mg/kg/
dose (up to a
maximum dose
of 250mg)
6-hourly for
7days

TMP: 4mg/kg,
SMX: 20mg/kg
twice daily for
7days (maximum
160mg TMP and
800mg SMX
12-hourly)

500mg single
dose on day 1,
then 250mg
single dose
fordays 25

500mg twice
daily for 7days

Adults

(maximum
1 g/day)
Erythromycin:
250mg 6-hourly
for 7days;

TMP: 160mg
twice daily, SMX:
800mg twice
daily for 7days

Erythromycin
ethyl succinate
(EES): 400mg
6-hourly for
7days

* Trimethoprim-sulfmethoxazole
Preferred for this age; refer to (c) Pertussis in pregnancy and (d) Use in infants and
infantile hypertrophic pyloric stenosis below.
Please refer to (d) Use in infants and infantile hypertrophic pyloric stenosis below.

Cases should be excluded from, for example, childcare facilities and school,
until they have taken 5days of antibiotic treatment. All cases, both suspect and
confirmed, should be notified to the State/Territory public health authorities (see
Appendix 1, Contact details for Australian, State and Territory Government health
authorities and communicable disease control).

236 The Australian Immunisation Handbook 9th Edition

(ii) Management of contacts of cases


(a) Vaccination
Since a primary course of 3 or more injections is required to protect against
pertussis, infant vaccination cannot be effectively used to control an outbreak.
However, unvaccinated or partially vaccinated contacts up to their 8th birthday
should be offered DTPa-containing vaccines and older individuals a single dose
of dTpa (see Section 1.3.5, Catch-up).
Passive immunisation with normal human immunoglobulin has not been shown
to be effective in the prevention of pertussis.
(b) Chemoprophylaxis

Based on these principles, prophylaxis is recommended for the following highrisk contacts of pertussis cases:
All household members when the household includes any child <24months
of age who has received fewer than 3 effective doses of pertussis vaccine
(ie. commenced after 6weeks of age with at least a 4-week interval between
doses, and the last dose given at least 14days previously).
Any woman in the lastmonth of pregnancy, regardless of vaccination status
(see Pertussis in pregnancy below).
All other children and adults in the same care group if the case, regardless
of his/her vaccination status, attended childcare for more than 1 hour while
infectious and that care group includes 1 or more children <24months of age
who have received fewer than 3 effective doses of pertussis vaccine.
Healthcare staff, regardless of vaccination status, working in a maternity
hospital or newborn nursery. Chemoprophylaxis is not recommended
routinely for healthcare staff caring for older infected children or adults.
Where a case worked in a maternity ward or newborn nursery for more than
an hour while infectious, then all babies in that ward should receive antibiotics.

Pertussis 237

3.14 Pertussis

In the usual clinical setting of delayed presentation and imperfect compliance,


the benefit of chemoprophylaxis in preventing the secondary transmission
of pertussis is likely to be limited.49,55 In view of this, the well established
(mainly gastrointestinal) side effects of erythromycin and the cost of the newer
macrolides, the use of chemoprophylaxis for prevention of secondary cases
should be reserved for those settings where the benefit is greatest. These settings
are best defined by the chance of transmission and the high risk of severe
complications should transmission occur. Close contacts can be defined as those
who either live in the same household (but not occasional sleepover contacts
unless they too are at increased risk of severe disease), or work in or attend
the same institutional setting (eg. maternity hospital ward, newborn nursery,
childcare centre) as a case.

Antibiotic regimens for chemoprophylaxis are the same as for cases (Table 3.14.1
above). Antibiotics should be given only if commenced either within 21days
of the onset of any symptoms, or within 14days of the onset of the paroxysmal
cough in the case. Childcare contacts in the same room as the case, who are not
age-appropriately vaccinated, should be excluded from childcare until the expiry
of 14days from their last exposure to the infectious case, unless they have already
completed 5days of a recommended antibiotic treatment, in which case they may
return.
(c) Pertussis in pregnancy
Treatment of pregnant women with pertussis onset within amonth of delivery
is important for the prevention of neonatal pertussis and, if the onset is within
3weeks of delivery, their newborn babies should also be given antibiotic therapy
(Table 3.14.1). Erythyromycin use earlier in pregnancy has well documented
safety (Category A). There are only limited data on the use of azithromycin in
pregnancy (Category B1).
(d) Use in infants and infantile hypertrophic pyloric stenosis
Several studies have shown an increased risk of infantile hypertrophic pyloric
stenosis (IHPS) when erythromycin is given for prophylaxis following exposure
to pertussis, especially in the first 2weeks of life.56,57 While there are, as yet, no
data available on the effectiveness of azithromycin use in infants <1month of
age, published case series report fewer adverse events following azithromycin
use when compared with erythromycin and, to date, there have been no reports
of IHPS in infants following use of azithromycin, although the size and number
of these studies is limited.58,59 Therefore, on currently available evidence,
and because of the risks of severe pertussis in this age group, azithromycin
is preferred to erythromycin for treatment and prophylaxis in infants aged
<1month by US authorities. Azithromycin is available as a suspension and
approved for use in Australia, but treatment and prophylaxis of pertussis are not
currently referred to in the product information. Parents of newborns prescribed
either erythromycin or azithromycin should be informed about the possible risks
for IHPS and counselled about signs of developing IHPS.

Use in pregnancy
Refer to Chapter 2.3, Groups with special vaccination requirements, Table 2.3.1
Vaccinations in pregnancy.

Variations from product information


The product information for both Infanrix hexa and Infanrix Penta states that
these vaccines may be given as a booster dose at 18months of age. NHMRC
recommends that a booster dose of DTPa (or DTPa-containing vaccines) is not
necessary at 18months of age. However, DTPa-containing vaccine may be used
for catch-up of the primary schedule in children <8years of age.

238 The Australian Immunisation Handbook 9th Edition

The product information for Infanrix-IPV states that this vaccine may be used as
a booster dose for children 6years of age who have previously been vaccinated
against diphtheria, tetanus, pertussis and poliomyelitis. NHMRC recommends
that booster doses of DTPa and IPV be given at 4years of age; however, this
product may be used for catch-up of the primary schedule or as a booster in
children <8years of age.
The product information for adolescent/adult formulations of dTpa-containing
vaccines states that these vaccines are indicated for booster doses only. NHMRC
recommends that, when a 3-dose primary course of diphtheria/tetanus toxoids
is given to an adolescent/adult, that dTpa replace the first dose of dT, with 2
subsequent doses of dT. If dT is not available, dTpa can be used for all 3 primary
doses, but this is not routinely recommended.

The product information for both clarithromycin and azithromycin do not list
the treatment or prophylaxis of pertussis as an approved indication for either
antibiotic. NHMRC recommends that these antibiotics may be used for the
treatment or prophylaxis of pertussis as per Table 3.14.1 above.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

Pertussis 239

3.14 Pertussis

The product information for Adacel and Boostrix (adolescent/adult formulations


of dTpa) states that these vaccines are recommended for use in those aged
>10years. However, NHMRC recommends that they may be used in people
aged 8years. The product information also states that dTpa should not be
given within 5years of a tetanus toxoid-containing vaccine. However, NHMRC
recommends that a single dose of dTpa vaccine can be administered at any time
following receipt of a diphtheria and tetanus toxoid-containing vaccine.

3.15 Pneumococcal disease


Bacteriology
Streptococcus pneumoniae are lancet shaped Gram-positive streptococci. To date,
90 capsular antigenic types have been recognised, each eliciting type-specific
immunity. Some of these types are commonly carried in the upper respiratory
tract, and some are more frequently associated with invasive disease. The
emergence of antibiotic-resistant strains of this organism has become an
increasing challenge with 2004 Australian data indicating that up to 18% of
invasive strains are resistant to 2 or more classes of antibiotics.1

Clinical features
Invasive pneumococcal disease (IPD) is defined as isolation of S. pneumoniae from
a normally sterile site, most commonly blood. The major clinical syndromes of
IPD include pneumonia, meningitis and bacteraemia without focus. In adults,
pneumococcal pneumonia is the most common clinical presentation of IPD,
while, in children, bacteraemia accounts for more than two-thirds of cases.2-4
The risk of IPD is highest in patients who cannot mount an adequate immune
response to pneumococcal capsular antigens, including those with functional
or anatomical asplenia, immunoglobulin deficiency, acute nephrotic syndrome,
multiple myeloma, HIV/AIDS, chronic renal failure, organ transplantation
and lymphoid malignancies.3,4 Other groups of patients, although generally
immunocompetent, develop IPD of higher incidence and/or severity. These
include people with chronic cardiovascular or pulmonary disease, diabetes
mellitus, alcohol-related problems, cirrhosis, or CSF leak after cranial trauma
or surgery, and those who smoke.3,5 In those without predisposing medical
conditions, both frequent otitis media and recently commencing childcare are
associated with increased risk of IPD in children,6 and tobacco smoking with
increased risk in adults.

Epidemiology
The highest rates of IPD are seen in children <2years of age and adults >85years
of age. In 2004, 2375 cases of IPD were notified to the National Notifiable
Diseases Surveillance System, a notification rate of 11.8 per 100000 population.1
The overall rate of IPD in Indigenous Australians was 3.2 times that in nonIndigenous Australians.1 In 2004, after implementation of the pneumococcal
conjugate vaccine program for high-risk children in 2001, the rate of IPD in
children <2years of age had decreased in Indigenous children (91.5 cases per
100000) to become similar to their non-Indigenous peers (93.6 cases per 100000).1
In the less-developed world and in some groups of Aboriginal and Torres Strait
Islander people, the incidence of IPD is as high as 200 per 100000 per year.

240 The Australian Immunisation Handbook 9th Edition

However, mortality rates among Indigenous Australian people are comparable


to those in non-Indigenous people, even in remote areas. Most non-Indigenous
adults who develop IPD have at least 1 risk factor, while most cases occurring
in Indigenous adults are associated with multiple risk factors. In adults, most
IPD isolates belong to serotypes contained in the 23-valent pneumococcal
polysaccharide vaccine.7-9
Among Indigenous children in northern Australia, before the introduction of the
7-valent pneumococcal conjugate vaccine, only about one-half to two-thirds of
IPD was caused by serotypes in the 7-valent pneumococcal conjugate vaccine
compared with 85% or more among non-Indigenous children.7,8,10 Nevertheless,
in north Queensland, a decrease in the annual incidence of IPD in the <5years
age group from 170 to 78 cases per 100000 was documented in the 3years after
introduction of the 7-valent pneumococcal conjugate vaccine.11 Similarly, the
annual incidence of vaccine-preventable IPD in Indigenous adults has declined
by 86% since the 23-valent pneumococcal polysaccharide vaccine was introduced
to north Queensland in 1986.11

Vaccines

Pneumococcal conjugate vaccine, 7-valent (7vPCV)


Prevenar Wyeth (7-valent pneumococcal conjugate vaccine; 7vPCV). Each
0.5mL monodose pre-filled syringe contains 2g of pneumococcal serotypes
4, 9V, 14, 18C, 19F, 23F and 4g of serotype 6B, conjugated to a mutant
non-toxic diphtheria toxin (CRM197) carrier protein, adsorbed onto 0.5mg
aluminium phosphate. Available in packs of 10 monodose pre-filled syringes.
7vPCV is approved for use in infants and children aged 6weeks to 9years.
Efficacy data from a pivotal trial in California found greater than 95% protective
efficacy against IPD due to the serotypes contained in the vaccine.12 A Cochrane
review of 4 trials assessing the efficacy of 7vPCV in children <2years of age found
7vPCV to be effective in reducing the incidence of IPD due to all serotypes, the
greater effect being seen in the reduction of IPD due to vaccine-related serotypes.13

Pneumococcal disease 241

3.15 Pneumococcal disease

There are currently 2 different types of pneumococcal vaccine available in


Australia. A 7-valent pneumococcal conjugate vaccine (7vPCV) became
available in 2001 for immunisation of infants and children aged from 6weeks
to 9years. 7vPCV was added to the NIP for high-risk children in 2001 and for
all children up to 2years of age from January 2005. The 23-valent pneumococcal
polysaccharide vaccine (23vPPV) has been available since 1983. A funded
program with 23vPPV for Indigenous Australians aged 50years began in
1999. Non-Indigenous Australians aged 65years became eligible to receive the
vaccine under the NIP from January 2005. In addition, people aged <65years
with underlying chronic conditions predisposing them to IPD can access 23vPPV
through the PBS.

Other pneumococcal infections in children (pneumonia and otitis media), not


associated with a positive sterile site culture, are also reduced by 7vPCV, but the
estimated reduction varies with case definition and severity. For clinically defined
otitis media or pneumonia, the reduction is similar at approximately 5%.14,15
A post-licensure study of 157471 children in California showed evidence of
disease reduction in unimmunised people, confirmed by a larger US study
showing a decline in the incidence of IPD of 52% in those 2039years of age and
26% in those 60years of age.16-18

Pneumococcal polysaccharide vaccine, 23-valent (23vPPV)


Pneumovax 23 CSL Biotherapies/Merck & Co Inc (23-valent pneumococcal
polysaccharide vaccine; 23vPPV). Each 0.5mL monodose vial contains 25g
of each of pneumococcal serotypes 1, 2, 3, 4, 5, 6B, 7F, 8, 9N, 9V, 10A, 11A,
12F, 14, 15B, 17F, 18C, 19F, 19A, 20, 22F, 23F and 33F; 0.25% phenol.
23vPPV contains polysaccharides derived from the 23 most frequent or most
virulent capsular types of S. pneumoniae in the USA. These same serotypes are
responsible for most IPD cases in adults in Australia. At least 90% of healthy
adults respond to the vaccine, with a 4-fold rise in type-specific antibody within
2 to 3weeks. Response to vaccine is diminished in patients with impaired
immunity and, in children <2years of age, is limited to a small number of
serotypes unless there has been previous 7vPCV vaccination.19
In developing countries with high attack rates, controlled trials have shown
that pneumococcal polysaccharide vaccine reduces mortality from pneumonia
in younger adults which, in this setting, is very likely to be pneumococcal.
Among at-risk individuals in developed countries with much lower attack rates,
a Cochrane review examining vaccines for preventing pneumococcal disease
reported that 23vPPV was effective in reducing the incidence of IPD, but not
non-bacteraemic pneumonia, among adults and the immunocompetent elderly.20
Similarly, a recent retrospective study in a managed care setting in the USA studied
47365 adults >65years of age over 3years, of whom 1428 were hospitalised with
community-acquired pneumonia and 61 developed documented pneumococcal
bacteraemia. Receipt of 23vPPV vaccine was associated with a significant reduction
in pneumococcal bacteraemia but not in hospitalisation for non-bacteraemic
pneumonia.21 In Australia, Victoria introduced a publicly funded 23vPPV program
for the >65years age group in 1998, resulting in an estimated 36% reduction in the
incidence of IPD and vaccine effectiveness of 71% (95% CI: 5482%).22

Transport, storage and handling


7-valent pneumococcal conjugate vaccine
Transport according to National Vaccine Storage Guidelines: Strive for 5.23 Store at
+2C to +8C. Do not freeze. Protect from light.

242 The Australian Immunisation Handbook 9th Edition

23-valent pneumococcal polysaccharide vaccine


Transport according to National Vaccine Storage Guidelines: Strive for 5.23 Store at
+2C to +8C. Do not freeze. Protect from light.

Dosage and administration


7-valent pneumococcal conjugate vaccine
The dose is 0.5mL by IM injection in the opposite limb to other injectable
vaccines if possible.

23-valent pneumococcal polysaccharide vaccine


The dose is 0.5mL as a single dose, by either SC or IM injection, in the opposite
limb to other injectable vaccines if possible.

Recommendations
7-valent pneumococcal conjugate vaccine
Vaccination of children
7vPCV is recommended in the NIP for all infants from 2months of age with a
catch-up for children up to 2years of age.
7vPCV may be safely given at the same time as other vaccines listed on the NIP
but must be administered using a separate injection site and limb.
7vPCV should be administered in a primary series of 3 doses at 2, 4 and 6months
of age. Unless there is an increased risk of IPD (see below), the additional benefits
are not considered sufficient to justify a routine (fourth) booster dose. This
recommendation is based on data from the pivotal randomised controlled trial
suggesting similar efficacy against type-specific IPD with either 3 or 4 doses.12
Subsequent studies from the UK examining immunogenicity data24 and the US
examining vaccine effectiveness25 were consistent with significant protection after
2 or more doses. The US study found higher vaccine effectiveness among those
who had received a fourth dose at 12months of age, but this was not statistically
significant.25 The current Australian dosing regimen will be regularly reviewed in
the light of trends in Australian IPD data and emerging international experience.

(ii) Aboriginal and Torres Strait Islander children living in the Northern
Territory, Queensland, South Australia and Western Australia
7vPCV should be administered at 2, 4 and 6months of age, followed at
1824months of age by a dose of 23vPPV. This recommendation is based on: (i) data
from several Australian studies which showed lower serotype coverage from the
7vPCV in similar populations;7,8,10 (ii) 2 large studies which demonstrated adequate
boosting responses to serotypes contained in the 7vPCV following 23vPPV;26,27
and (iii) 2 large studies which demonstrated adequate primary responses to some
serotypes in 23vPPV, but not in 7vPCV, from 1824months of age.28,29

Pneumococcal disease 243

3.15 Pneumococcal disease

(i) Healthy children

(iii) Children with underlying medical conditions (listed in Table


3.15.2) associated with greater risk or severity of IPD
7vPCV should be administered at 2, 4 and 6months of age, followed by a fourth
dose of 7vPCV at 12months of age and a booster dose of 23vPPV at 45years of
age. This is based on data showing lower immune responses in these children to
certain serotypes in 7vPCV which can be enhanced by an additional dose, and
their continuing susceptibility to IPD at older ages, with a higher prevalence of
serotypes not contained in the 7vPCV.30

(iv) Children with asplenia (functional or anatomical)


9years of age (ie. before their 10th birthday)
with no previous history of pneumococcal vaccination with 7vPCV or
23vPPV
2 doses of 7vPCV given 2months apart, followed by 23vPPV at least
2months after the last dose of 7vPCV. This is based on an inadequate
response to 1 dose of 7vPCV among some asplenic individuals which is
enhanced by a second dose.27
with previous history of pneumococcal vaccination with 7vPCV or 23vPPV
where a dose of 23vPPV was given more than 6months ago but no doses
of 7vPCV have been administered, give 2 doses of 7vPCV at least 2months
apart. This is based on inadequate response to 1 dose of 7vPCV in some
asplenic individuals which seems unlikely to be influenced by previous
receipt of 23vPPV, although no specific data are available.
where previous doses of 7vPCV have been administered but no 23vPPV,
give 23vPPV at least 2months after the last 7vPCV dose. This is based on
similar considerations to those above.
NB. Children with asplenia should also be considered for other interventions (see
Chapter 2.3, Subsection 2.3.3.5, Individuals with functional or anatomical asplenia).

(v) Children 9years of age (ie. before their 10th birthday) who have been
diagnosed with an underlying medical condition (listed in Table 3.15.2 below)
after they received the infant schedule of 7vPCV at 2, 4 and 6months of age
Where a previously healthy child, currently aged >12months, was vaccinated
according to the NIP schedule and received 7vPCV at 2, 4 and 6months of age
but has since developed or been diagnosed with a condition listed in Table 3.15.2
below, he/she should receive a further dose of 7vPCV followed 2months later by
a dose of 23vPPV.

244 The Australian Immunisation Handbook 9th Edition

Table 3.15.1: Summary table pneumococcal vaccination schedule for children


9years of age (see also Section 1.3.5, Catch-up)
For childhood immunisation schedule (children 5years of age)
7vPCV

23vPPV

Comments

All healthy children


(including Indigenous
children residing
in ACT, NSW,
TAS and VIC)

2, 4 and
6months of
age (up to
2years of age)*

No

If delays in start of schedule


after 2months, refer Section
1.3.5, Catch-up, Table 1.3.9.

Indigenous children
residing in NT, QLD,
SA and WA only

2, 4 and
6months of
age (up to
2years of age)

1824months

If delays in start of schedule


after 2months, refer Section
1.3.5, Catch-up, Table 1.3.10.

Children with
underlying medical
conditions (refer
Table 3.15.2)

2, 4, 6 and
12months
of age

45years

If delays in start of schedule


after 2months, refer Section
1.3.5, Catch-up, Table 1.3.11.

For children 69years of age with underlying medical conditions as listed in Table
3.15.2
7vPCV

23vPPV

Comments

2 doses, at least Give 1 dose at least


2months apart 2months after last
dose of 7vPCV

For revaccination schedules


for children 10years, refer
to Table 3.15.3 below.

Has received 7vPCV


primary course at 2, 4
and 6months of age

1 dose

Give 1 dose at least


2months after last
dose of 7vPCV

For revaccination schedules


for children 10years, refer
to Table 3.15.3 below.

History of at least
2 7vPCV doses,
and no 23vPPV

1 dose

Give 1 dose at least


2months after last
dose of 7vPCV

For revaccination schedules


for children 10years, refer
to Table 3.15.3 below.

History of 23vPPV,
but no 7vPCV

Give 2 doses at Refer revaccination


least 2months schedule Table 3.15.3
apart, starting
for further schedules
at least
6months after
dose of 23vPPV

For revaccination schedules


for children 10years, refer
to Table 3.15.3 below.

* Immunisation of healthy children (including Indigenous children residing in ACT, NSW,


VIC, and TAS) only up to 2years of age.

Pneumococcal disease 245

3.15 Pneumococcal disease

No history of any
pneumococcal
vaccination

Booster doses of 7vPCV


With the exception of children with underlying medical conditions (see above),
booster doses of 7vPCV are not required.
For details of catch-up schedules, please refer to Section 1.3.5, Catch up.
Table 3.15.2: Underlying medical conditions predisposing children 9years of
age to IPD
Diseases compromising immune response to pneumococcal infection:
congenital immune deficiency including symptomatic IgG subclass
or isolated IgA deficiency (but children who requiremonthly
immunoglobulin infusion are unlikely to benefit from vaccination),
immunosuppressive therapy (including corticosteroid therapy 2mg/kg per day
of prednisolone or equivalent for more than 2weeks) or radiation therapy, where
there is sufficient immune reconstitution for vaccine response to be expected,
compromised splenic function due to sickle haemoglobinopathies,
or congenital or acquired asplenia,
haematological malignancies,
HIV infection, before and after development of AIDS,
renal failure, or relapsing or persistent nephrotic syndrome,
Down syndrome.
Anatomical or metabolic abnormalities associated with higher rates or severity of IPD:
cardiac disease associated with cyanosis or cardiac failure,
all premature infants with chronic lung disease,
all infants born at less than 28weeks gestation,
cystic fibrosis,
insulin-dependent diabetes mellitus,
proven or presumptive cerebrospinal fluid (CSF) leak,
intracranial shunts and cochlear implants.

23-valent pneumococcal polysaccharide vaccine


23vPPV may be safely given at the same time as other vaccines listed on the NIP
but must be administered using a separate injection site and limb.

(i) 23vPPV is recommended for:


All people aged 65years.
Aboriginal and Torres Strait Islander people 50years of age and those
1549years of age who have underlying conditions placing them at risk of IPD.
People aged 10years who have underlying chronic illnesses predisposing
them to IPD including:

246 The Australian Immunisation Handbook 9th Edition

asplenia either functional (including sickle-cell disease) or anatomical;


where possible, the vaccine should be given at least 14days before
splenectomy,
conditions associated with increased risk of IPD due to impaired
immunity, eg. HIV infection before the development of AIDS, acute
nephrotic syndrome, multiple myeloma, lymphoma, Hodgkins disease
and organ transplantation,
chronic illness associated with increased risk of IPD including chronic
cardiac, renal or pulmonary disease, diabetes, alcohol-related problems,
CSF leak.
Tobacco smokers.

(ii) 23vPPV booster dose is recommended following previous 7vPCV


At 1824months of age, after a primary series of 7vPCV, in Aboriginal and
Torres Strait Islander children in the Northern Territory, Queensland, South
Australia and Western Australia (see Section 1.3.5, Catch up, Table 1.3.10).
At 45years of age in children at risk of either high incidence or severity of
IPD because of underlying medical conditions (see Table 3.15.2), following a
primary series of 7vPCV (see Section 1.3.5, Catch up, Table 1.3.11).

iii) Revaccination with 23vPPV

Although an early study raised concerns about extensive local adverse events
following revaccination with 23vPPV,31 several recent studies have shown that 3
doses (ie. 2 revaccinations) of 23vPPV are not associated with more local adverse
events compared to 1 or 2 doses.32,33
Less clear, however, is the adequacy of the immune response after revaccination
with 23vPPV. Although an earlier study reported that there was an immune
hyporesponsiveness after a first revaccination, more recent studies suggest that
the immune responses to revaccination may be adequate.31,34

Pneumococcal disease 247

3.15 Pneumococcal disease

A maximum of 3 doses (ie. 2 revaccinations) of 23vPPV are recommended, based


on data concerning adverse events and effectiveness.

Table 3.15.3: Revaccination with 23vPPV for people 10years of age


Primary dose 23vPPV
given to

First 23vPPV revaccination

Second 23vPPV
revaccination

Non-Indigenous
adults 65years

5years after first dose

No

Non-Indigenous
adults <65years with
underlying chronic medical
condition or smoker

5years after first dose

Either 5years after first


revaccination or at 65years
of age (whichever is later)

Indigenous adults
aged 50years

5years after first dose

No

Indigenous adults
aged <50years with
underlying chronic medical
condition or smoker

5years after first dose

Either 5years after first


revaccination or at 50years
of age (whichever is later)

Asplenic individuals

5years after first dose

Either 5years after first


revaccination or at 50years
of age (for Indigenous
adults) or 65years of
age (for non-Indigenous
adults), whichever is later

NB. Indigenous children in the Northern Territory, Queensland, South


Australia and WesternAustralia receive 23vPPV at 1824months of age (see
Recommendations, point (ii) above). This childhood dose is not relevant to the
recommendations concerning revaccination given in Table3.15.3.

Contraindications
7-valent pneumococcal conjugate vaccine
The only absolute contraindications to 7vPCV are:
anaphylaxis following a previous dose of the vaccine, or
anaphylaxis following any vaccine component.

23-valent pneumococcal polysaccharide vaccine


The only absolute contraindications to 23vPPV are:
anaphylaxis following a previous dose of the vaccine, or
anaphylaxis following any vaccine component.

248 The Australian Immunisation Handbook 9th Edition

Relative contraindications include the following:


Age <2years the immune response in young children is restricted to a few
serotypes (so benefits of immunisation are limited) unless previously given 1
or more doses of 7vPCV.
Recent use of immunosuppressive therapy or radiation of lymph nodes.
However, once it is considered that these patients are immunologically
stabilised, they should be promptly vaccinated.

Adverse events
7-valent pneumococcal conjugate vaccine
Among the most commonly reported are injection site adverse events and fever.
7vPCV is more commonly associated with local adverse events, with rates of
erythema ranging from 10.0 to 11.6% (very common) for 7vPCV, compared
with 6.7 to 11.4% (common to very common) for DTPa. There is no pattern
of increasing local reactogenicity with subsequent doses.12 A higher rate of
local adverse events has been observed in older children after a single dose.
Prophylactic antipyretic medication is recommended in children who have
seizure disorders or a previous history of febrile seizures.

23-valent pneumococcal polysaccharide vaccine

Previously, there were concerns about extensive local adverse events following
revaccination with 23vPPV31 but recent studies indicate that revaccination is
not associated with more local adverse events compared to 1 or 2 doses.32,33
Revaccination is not associated with an increase in systemic adverse events such
as fever or headache.32-34

Use in pregnancy
7-valent pneumococcal conjugate vaccine
Vaccination during pregnancy has not been evaluated for potential harmful
effects in animals or humans. Although unlikely to result in adverse effects to
mother or fetus, it is neither indicated nor recommended.

23-valent pneumococcal polysaccharide vaccine


Although 23vPPV has been administered in pregnancy in the context of clinical
trials with no evidence of adverse effects, data are limited and deferral of
vaccination is recommended unless there is an increased risk of IPD.35,36 Women
of reproductive age with known risk factors for IPD should be vaccinated before
planned pregnancy.

Pneumococcal disease 249

3.15 Pneumococcal disease

About half the recipients of 23vPPV will experience some soreness after the first
dose, but pain or swelling severe enough to limit arm movement occurs in less
than 5% (common) of recipients.31 Low-grade fever occurs occasionally, but fever
above 39C occurs in less than 0.5% (uncommon) of recipients.31

Variations from product information


7-valent pneumococcal conjugate vaccine
The product information recommends a 4-dose 7vPCV schedule for vaccination
commencing at 2months of age with doses at 2, 4, 6 and 12months of age,
3 doses for vaccination commencing between 7 and 12months of age, and 2
doses for vaccination commencing between 13 and 23months of age. However,
NHMRC recommends 1 dose less than that stated in the product information for
healthy children who are not at increased risk of IPD.

23-valent pneumococcal polysaccharide vaccine


23vPPV is licensed for use only in children >24months of age, but NHMRC
considers that it can be used from 18months of age in children who have
previously received 7vPCV.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

250 The Australian Immunisation Handbook 9th Edition

3.16 Poliomyelitis
Virology
Polioviruses are classified as enteroviruses in the family Picornaviridae.1 They
have an RNA genome, and are transient inhabitants of the gastrointestinal tract
(GIT). There are 3 poliovirus serotypes, PV1, PV2 and PV3. The virus enters
through the mouth, multiplies in the pharynx and GIT and is excreted in the
stools for severalweeks. The virus invades local lymphoid tissue, enters the
blood stream and may then infect and replicate in cells of the central nervous
system.2

Clinical features
Poliomyelitis is an acute illness following gastrointestinal infection by one of the
3 types of poliovirus. Transmission is through faecal-oral and, occasionally, oraloral spread.3 The infection may be clinically inapparent. If symptoms occur, they
may include headache, gastrointestinal disturbance, malaise and stiffness of the
neck and back, with or without paralysis. Paralysis is classically asymmetrical.
Paralytic polio is a complication of poliovirus aseptic meningitis, and may be
spinal (79%), bulbar (2%) or bulbospinal (19%). The case-fatality rate in paralytic
polio is 2 to 5% in children, 15 to 30% in adults and up to 75% in bulbar polio.
The infection rate in households with susceptible young children can reach 100%.
The proportion of inapparent or asymptomatic infection to paralytic infection
may be as high as 1000:1 in children and 75:1 in adults, depending on the
poliovirus type and social and environmental conditions.2
The incubation period ranges from 3 to 21days. Infected individuals are most
infectious from 7 to 10days before to 7 to 10days after the onset of symptoms.
The oral vaccine virus may be shed in the faeces for 6weeks or more,2 and for up
to severalyears in people with impaired immunity. Oral vaccine strains shed for
manyyears may mutate into potentially neurovirulent strains.4-9

Epidemiology

Poliomyelitis 251

3.16 Poliomyelitis

The incidence of poliomyelitis has been dramatically reduced worldwide, but


cases still occur in developing countries in the Indian subcontinent, the eastern
Mediterranean and Africa.10,11 The World Health Organization (WHO) aimed
to eradicate poliomyelitis by the year 2005 and, although not successful, is still
hopeful this will be achieved by 2010 or soon after.12 In 1994, the continents of
North and South America were certified to be free of polio,13 followed by the
Western Pacific region (including Australia) in 2000 and the European region
in 2002.14,15 In countries where the disease incidence is low but transmission is
still occurring, poliomyelitis cases are seen sporadically or as outbreaks among
non-vaccinated individuals. In 2005, 12 countries previously declared polio-

free, including Indonesia, experienced outbreaks due to importations of wild


poliovirus from one of the remaining endemic countries: Afghanistan, India,
Nigeria and Pakistan.11
In Australia, the peak incidence of poliomyelitis was 39.1/100000 in 1938.
There has been a dramatic fall in incidence since 1952, but epidemics occurred
in 1956 and 196162. The last notified case of wild poliomyelitis in Australia
occurred in 1977 due to an importation from Turkey, but 2 vaccine-associated
cases were notified in 1986 and 1995.16,17 Because of the rapid progress in
global polio eradication and diminished risk of wild virus associated disease,
inactivated poliomyelitis vaccine (IPV) is now used for all doses of polio vaccine
in Australia.3,18 This change was implemented because of concern about the risk of
causing vaccine-associated paralytic poliomyelitis (VAPP), which is about 1 case
for every 2.4 million doses of oral poliomyelitis vaccine (OPV) distributed.19 The
advantage of using IPV is that it cannot cause VAPP.

Global eradication of polio


The WHO strongly supports the use of OPV to achieve global eradication of
poliomyelitis, especially in countries with continued or recent circulation of wildtype poliovirus.20 However, most countries which can afford IPV now use IPV
in preference to OPV, in order to eliminate the risk of VAPP and also to reduce
the risk of prolonged shedding of potentially neurovirulent strains of poliovirus
by individuals with impaired immunity.3 A vaccine-derived poliovirus (VDPV)
is derived from OPV but has a number of significant mutations due to longterm replication in an individual with impaired immunity (iVDPV) or through
person-to-person transmission in areas of low polio vaccine coverage (circulating
VDPV or cVDPV). Outbreaks of poliomyelitis due to cVDPV have been reported
worldwide.6 People travelling to countries still using OPV are at risk of VAPP,
as was reported for an unimmunised adult from the USA who travelled to
Costa Rica in 2005.21 The WHO is planning for global OPV cessation, once the
interruption of wild poliovirus transmission has been certified, to remove the
incidence of VAPP and VDPVs.22 Further information is available from the WHO
Polio Eradication website http://www.polioeradication.org.

252 The Australian Immunisation Handbook 9th Edition

Vaccines
IPOL Sanofi Pasteur Pty Ltd (IPV; inactivated poliomyelitis vaccine). Each
0.5mL pre-filled syringe contains poliovirus 40D antigen units of type 1, 8D
antigen units of type 2 and 32D antigen units of type 3 grown on monkey
kidney cells, inactivated with formaldehyde; traces of phenoxyethanol as
preservative, neomycin, streptomycin and polymyxin B.
Combination vaccines that include IPV
Formulations for children aged <8years
Infanrix hexa GlaxoSmithKline (DTPa-hepB-IPV-Hib; diphtheriatetanus-acellular pertussis-hepatitis B-inactivated poliomyelitis vaccineHaemophilus influenzae type b (Hib)). The vaccine consists of both a 0.5mL
pre-filled syringe containing 30 IU diphtheria toxoid, 40 IU tetanus toxoid,
25g pertussis toxoid (PT), 25g filamentous haemagglutinin (FHA), 8g
pertactin (PRN), 10g recombinant HBsAg, 40 D-antigen units inactivated
polioviruses type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32
D-antigen units type 3 (Saukett) adsorbed onto aluminium hydroxide/
phosphate; phenoxyethanol as preservative; traces of formaldehyde,
polymyxin and neomycin and a vial containing a lyophilised pellet of 10g
purified Hib capsular polysaccharide (PRP) conjugated to 2040g tetanus
toxoid. The vaccine must be reconstituted by adding the entire contents of the
syringe to the vial and shaking until the pellet is completely dissolved. May
also contain yeast proteins.
Infanrix-IPV GlaxoSmithKline (DTPa-IPV; diphtheria-tetanus-acellular
pertussis-inactivated poliomyelitis vaccine). Each 0.5mL pre-filled syringe
contains 30 IU diphtheria toxoid, 40 IU tetanus toxoid, 25g PT, 25g FHA,
8g PRN, 40 D-antigen units inactivated polioviruses type 1 (Mahoney),
8 D-antigen units type 2 (MEF-1) and 32 D-antigen units type 3 (Saukett)
adsorbed onto aluminium hydroxide; phenoxyethanol as preservative;
traces of formaldehyde, polymyxin and neomycin.

Poliomyelitis 253

3.16 Poliomyelitis

Infanrix Penta GlaxoSmithKline (DTPa-hepB-IPV; diphtheria-tetanusacellular pertussis-hepatitis B-inactivated poliomyelitis vaccine). Each
0.5mL pre-filled syringe contains 30 IU diphtheria toxoid, 40 IU tetanus
toxoid, 25g PT, 25g FHA, 8g PRN, 10g recombinant HBsAg, 40
D-antigen units inactivated polioviruses type 1 (Mahoney), 8 D-antigen
units type 2 (MEF-1) and 32 D-antigen units type 3 (Saukett) adsorbed
onto aluminium hydroxide/phosphate; phenoxyethanol as preservative;
traces of formaldehyde, polymyxin and neomycin. May also contain
yeast proteins.

Formulations for people aged 8years


Adacel Polio Sanofi Pasteur Pty Ltd (dTpa; diphtheria-tetanus-acellular
pertussis- inactivated poliomyelitis vaccine). Each 0.5mL monodose vial
contains 2 IU diphtheria toxoid, 20 IU tetanus toxoid, 2.5g PT, 5g
FHA, 3g PRN, 5g pertussis fimbriae (FIM) 2+3; 40 D-antigen units
inactivated polioviruses type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1)
and 32 D-antigen units type 3 (Saukett); 1.5mg aluminium phosphate;
phenoxyethanol as preservative; traces of formaldehyde, polymyxin,
neomycin and streptomycin.
Boostrix-IPV GlaxoSmithKline (dTpa-IPV; diphtheria-tetanus-acellular
pertussis-inactivated poliomyelitis vaccine). Each 0.5mL pre-filled syringe
contains 2 IU diphtheria toxoid, 20 IU tetanus toxoid, 8g PT, 8g FHA,
2.5g PRN, 40 D-antigen units inactivated polioviruses type 1 (Mahoney),
8 D-antigen units type 2 (MEF-1) and 32 D-antigen units type 3 (Saukett)
adsorbed onto aluminium hydroxide/phosphate; traces of formaldehyde,
polymyxin and neomycin.
IPV (IPOL) and IPV-containing combination vaccines contain polioviruses of
types 1, 2 and 3 inactivated by formaldehyde. A course of 3 injections with an
interval of 2months between each dose produces long-lasting immunity (both
mucosal and humoral) to all 3 poliovirus types. IPV produces considerably lower
levels of intestinal immunity than OPV.

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.23 Store at
+2C to +8C. Do not freeze. Protect from light.

Dosage and administration


The dose of IPV (IPOL) and of the IPV-containing combination vaccines is
0.5mL. IPV is given by SC injection, whereas the IPV-containing vaccines
are administered by IM injection. If IPV (IPOL) is inadvertently given
intramuscularly, there is no need to repeat the dose.

Recommendations
Primary vaccination of infants and children
(i) IPV (IPOL) or IPV-containing vaccines are recommended for infants from
2months of age. An open, randomised, multi-centre trial comparing the
hexavalent and pentavalent IPV-containing vaccines found that infants receiving
either vaccine at 2, 4 and 6months of age had seroprotective levels of antibody to
polio virus types 1, 2 and 3.24

254 The Australian Immunisation Handbook 9th Edition

Extra doses of IPV (IPOL) or IPV-containing vaccines are not needed for babies
born prematurely.
(ii) The primary course consists of 3 separate doses of vaccine. An interval of
2months between each dose is recommended, but the minimum interval can be
as short as 1month for catch-up.
(iii) Interchangeability of OPV and IPV: Oral poliomyelitis vaccine (OPV)
is no longer in use in Australia. OPV and IPV are interchangeable. Children
commenced on OPV should complete their polio vaccination schedule using IPV
(IPOL) or IPV-containing vaccines.

Primary vaccination of adults


A course of 3 doses of IPV (IPOL) or IPV-containing vaccines at intervals of 1 to
2months is recommended for the primary vaccination of adults. No adult should
remain unvaccinated against poliomyelitis.

Booster doses
Children
A booster dose of IPV (IPOL) or IPV-containing vaccine should be given at
4years of age. A fifth dose of IPV is no longer recommended as Australia has
been declared polio free since 200014 and, as in the US, a completed poliomyelitis
vaccination schedule for children is 3 primary doses and 1 booster dose of IPV
(IPOL) or an IPV-containing vaccine.25
Adults
Booster doses for adults are not necessary unless they are at special risk, such as:
travellers to areas or countries where poliomyelitis is epidemic or endemic
(see http://www.polioeradication.org for more information on affected
countries), or
healthcare workers, including laboratory workers, in possible contact with
poliomyelitis cases.
For those exposed to a continuing risk of infection, booster doses are desirable every
10years. dTpa-IPV combination vaccines can be used where otherwise indicated.

Contraindications
anaphylaxis following a previous dose of the vaccine, or
anaphylaxis following any component of the vaccine.

Adverse events
IPV-containing vaccines cause erythema (33%, very common), pain (13%, very
common), and induration (1%, uncommon) at the injection site. Other symptoms
reported in young babies are: fever, crying and decreased appetite (510%, common).

Poliomyelitis 255

3.16 Poliomyelitis

The only absolute contraindications to IPV (IPOL) or IPV-containing vaccines are:

Use in pregnancy
Refer to Chapter 2.3, Groups with special vaccination requirements, Table 2.3.1
Vaccinations in pregnancy.

Variations from product information


The product information for IPV suggests that the fourth dose be given 12months
after the third dose for both adults and children, followed by a fifth dose for
children at 4years of age. NHMRC recommends the fourth dose for children at
4years of age and no fourth dose for adults unless they are at special risk.
The product information suggests that any sensitivity to vaccine components is a
contraindication, whereas NHMRC recommends that the only contraindication is
a history of anaphylaxis to a previous dose or to any of the vaccine components.
The product information for both Infanrix hexa and Infanrix Penta states that
these vaccines may be given as a booster dose at 18months of age. NHMRC
recommends that a booster dose of DTPa (or DTPa-containing vaccines) is not
necessary at 18months of age. However, DTPa-containing vaccine may be used
for catch-up of the primary schedule in children <8years of age.
The product information for Infanrix-IPV states that this vaccine may be used as
a booster dose for children 6years of age who have previously been vaccinated
against diphtheria, tetanus, pertussis and poliomyelitis. NHMRC recommends
that booster doses of DTPa and IPV be given at 4years of age; however, this
product may be used for catch-up of the primary schedule or as a booster in
children <8years of age.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

256 The Australian Immunisation Handbook 9th Edition

3.17Q fever

Q fever is caused by Coxiella burnetii, an obligate intracellular bacterium,


classified in a separate genus, Coxiella. A near relative is Legionella pneumophilia.1
The organism is slightly more resistant to heat than other vegetative bacteria,
but nevertheless is inactivated at pasteurisation temperatures. It survives well in
air, soil, water and dust and may also be disseminated on fomites such as wool,
hides, clothing, straw and packing materials.2,3

Clinical features
Q fever can be acute or chronic, and there is increasing recognition of long-term
sequelae. However, in many instances, infection can be asymptomatic.4,5
Acute Q fever usually has an incubation period of 2 to 3weeks, depending
on the inoculum size and other variables6 (range from 4days up to 6weeks).
Clinical symptoms vary by country but in Australia it commonly presents with
rapid onset of high fever, rigors, profuse sweats, extreme fatigue, muscle and
joint pain, severe headache and photophobia.4,5 As the attack progresses there is
usually evidence of hepatitis, occasionally with frank jaundice; a proportion of
patients may have pneumonia which is usually mild but can require mechanical
ventilation. If untreated, the acute illness lasts 1 to 3weeks and may be
accompanied by substantial weight loss in the more severe cases.4,5
C. burnetii may cause chronic manifestations, the most commonly reported being
subacute endocarditis. Less common presentations include granulomatous
lesions in bone, joints, liver, lung, testis and soft tissues. Infection in early
pregnancy, or even before conception, may recrudesce at term and cause fetal
damage.7-9
Recent studies have also identified a late sequel to infection, post Q fever fatigue
syndrome (QFS), which occurs in about 10 to 15% of patients with acute Q
fever.10-13 Laboratory research suggests that C. burnetii persists in most instances
of acute Q fever, regardless of clinical status, and that immunogenic variation
in the response to persistent infection leads to cytokine dysregulation and
determines whether QFS occurs.11,14,15

Epidemiology
C. burnetii infects both wild and domestic animals and their ticks, with cattle,
sheep and goats being the main source of human infection. Companion animals
such as cats and dogs may also be infected. The animals shed C. burnetii into the
environment through their products of conception (especially high numbers of
coxiellas) but also in their milk, urine, and faeces. C. burnetii is highly infectious16
and can survive in the environment. The organism is transmitted to humans via

Q fever 257

3.17Q fever

Bacteriology

the inhalation of infected aerosols or dust. Those most at risk include workers
from the meat and livestock industries and shearers, with non-immune new
employees or visitors being at highest risk of infection. Nevertheless, Q fever is
not confined to occupationally exposed groups; there are numerous reports of
sporadic cases or outbreaks in the general population in proximity to infected
animals in stockyards, feedlots, processing plants or farms.
Use of Q fever vaccine in Australia can be considered in 3 periods. First, from
1991 to 1993 when vaccine was used in a limited number of abattoirs, then from
1994 to 2000 when vaccination steadily increased to cover large abattoirs in
most states,17 and finally from 2001 to 2006 during the period of the Australian
Government sponsored Q fever Management Program.18 This program extended
vaccination to farmers, their families and employees in the livestock-rearing
industry. With respect to abattoir workers, there has been a clear reduction
in Q fever cases and associated insurance claims since 1994.17,19 More widely,
the numbers of Q fever cases reported to the National Notifiable Diseases
Surveillance System (NNDSS) have declined over the period 1994 to 2005,
during which there has been an increasing use of vaccine (see Figure 3.17.1). This
decline is suggestive of an impact from vaccination among people not working
in abbatoirs but, as there are substantial variations in total numbers of cases from
year to year, requires confirmation over a longer period.20
Figure 3.17.1: Q fever notifications and hospitalisations, Australia, 1993 to
2005,* bymonth of diagnosis or admission20
100

90

Notifications
Hospitalisations

80

70

Number

60

50

40

30

20

10

0
Jan Jul Jan Jul Jan Jul Jan Jul Jan Jul Jan Jul Jan Jul Jan Jul Jan Jul Jan Jul Jan Jul Jan Jul Jan Jul
1993 1993 1994 1994 1995 1995 1996 1996 1997 1997 1998 1998 1999 1999 2000 2000 2001 2001 2002 2002 2003 2003 2004 2004 2005 2005

* Notifications where themonth of diagnosis was between January 1993 and December 2005;
hospitalisations where themonth of admission was between 1 July 1993 and 30 June 2005.

258 The Australian Immunisation Handbook 9th Edition

Vaccine4,21

Q-VAX Skin Test CSL Biotherapies (Q fever skin test). Each 0.5mL liquid
vial when diluted in 15mL of sodium chloride contains 16.6 ng of purified
killed suspension of Coxiella burnetii in each diluted 0.1mL dose; thiomersal
0.01% w/v before dilution. May contain egg proteins.
Q fever vaccine and skin test consist of a purified killed suspension of C.
burnetii. It is prepared from the Phase I Henzerling strain of C. burnetii grown
in the yolk sacs of embryonated eggs. The organisms are extracted, inactivated
with formalin, and freed from excess egg proteins by fractionation and
ultracentrifugation. Thiomersal 0.01% w/v is added as a preservative.
Phase I whole-cell vaccines have been shown to be highly antigenic and
protective against challenge both in laboratory animals and in volunteer trials.22
Serological response to the vaccine is chiefly IgM antibody to C. burnetii Phase
I antigen. In subjects weakly seropositive before vaccination, the response
is mainly IgG antibody to Phase I and Phase II antigens.23 Although the
seroconversion rate may be low, long-term cell-mediated immunity develops24
and the vaccine has been shown to be protective in open and placebo-controlled
trials, and in 2 post-licensing trials, to have a vaccine efficacy of 100%.25-28 Lack of
seroconversion is not a reliable marker of lack of vaccination.22
During recentyears, with much larger numbers vaccinated, a few instances
of laboratory proven Q fever have been observed in vaccinated subjects.21 It is
important that these apparent vaccine failures are fully investigated and that
vaccination status is reported for all notified cases.
It should be noted that vaccination during the incubation period of a natural
attack of Q fever does not prevent the development of the disease.22
A useful website for Q fever vaccine providers is http://www.qfever.org/vaclist.php.

Transport, storage and handling


Transport the vaccine according to National Vaccine Storage Guidelines: Strive for
5.29 Store at +2C to +8C, and do not freeze or store in direct contact with ice
packs. If vaccine has been exposed to temperatures less than 0C, do not use.
Protect from light.

Q fever 259

3.17Q fever

Q-VAX CSL Biotherapies (Q fever vaccine). Each 0.5mL pre-filled syringe


contains 25g purified killed suspension of Coxiella burnetii; thiomersal
0.01% w/v. May contain egg proteins.

Dosage and administration


A single dose of 0.5mL of Q-VAX is given by SC injection after ascertaining that
serological and skin testing have been performed and that both tests are negative
(see Pre-vaccination testing below).

Recommendations
Q fever vaccine is recommended for those at risk of infection with C. burnetii.
This includes abattoir workers, farmers, stockyard workers, shearers, animal
transporters, and others exposed to cattle, camels, sheep, goats and kangaroos or
their products (including products of conception). It also includes veterinarians,
veterinary nurses, veterinary students, agricultural college staff and students
(working with high-risk animals) and laboratory personnel handling veterinary
specimens or working with the organism (see also Chapter 2.3, Groups with special
vaccination requirements, Table 2.3.6 Recommended vaccinations for those at risk of
occupationally acquired vaccine-preventable diseases).
Workers at pig abattoirs do not require Q fever vaccination.

Pre-vaccination testing
(i) Before vaccination, people with a negative history of previous Q fever must
have serum antibody estimations and skin tests to exclude those likely to have
hypersensitivity reactions to the vaccine resulting from previous (possibly
unrecognised) exposure to the organism.
(ii) If the person has a positive history of previous infection with Q fever, or has
already been vaccinated for Q fever, skin testing and serology are not required
and vaccination is contraindicated.
(iii) Note that a few subjects who have had verified Q fever in the past show no
response to serological or skin testing. However, such subjects may experience
serious reactions to administration of Q fever vaccine. Thus, it is vital to take a
detailed history and to obtain documentation of previous Q fever vaccination or
laboratory results confirming Q fever disease in all potential vaccinees; those who
have worked for more than 10years in the livestock or meat industries should be
questioned particularly carefully. If there is any doubt about serological results
or skin testing, they should be repeated 2 to 3weeks later (see (vi) below for
interpretation).
(iv) Antibody studies were originally done by complement fixation (CF) tests at
serum dilutions of 1 in 2.5, 5 and 10 against the Phase II antigen of C. burnetii.
Although this is generally satisfactory, many testing laboratories now use
enzyme immunoassay (EIA) or immunofluorescent antibody (IFA) to detect IgG
antibody to C. burnetii as an indicator of past exposure. Subjects CF antibody
positive at 1 in 2.5, IFA positive at 1 in 10 or more, or with a definite positive
absorbance value in the EIA, should not be vaccinated (see Table 3.17.1).

260 The Australian Immunisation Handbook 9th Edition

Table 3.17.1: Interpretation and action for serological and skin test results (with
modifications from Q fever. Your questions answered (CSL, 1999)4)
Serology

Skin test

Interpretation/Action

Positive antibody test*

Positive

Sensitised: do not vaccinate

Equivocal antibody test^

Negative antibody test

Borderline

Sensitised: do not vaccinate

Negative

Sensitised: do not vaccinate

Positive

Sensitised: do not vaccinate

Borderline

Indeterminate (see (vi) below)

Negative

Indeterminate (see (vi) below)

Positive

Sensitised: do not vaccinate

Borderline

Indeterminate (see (vi) below)

Negative

Non-immune: vaccinate

* Positive antibody test: CF antibody or IFA positive (according to criteria used by


diagnosing laboratory); or definite positive EIA absorbance value (according to
manufacturers instructions).
Positive skin test: induration present.
Borderline skin test: induration just palpable.
Negative skin test: no induration.
^ Equivocal antibody test: CF antibody or IFA equivocal (according to criteria used by
diagnosing laboratory); or equivocal EIA absorbance value (according to manufacturers
instructions).
# Negative antibody test: CF antibody or IFA negative (according to criteria used
by diagnosing laboratory); or definite negative EIA absorbance value (according to
manufacturers instructions).

(vi) Test results are indeterminate when skin test induration is just palpable and/
or there is an equivocal level of antibodies in one or other of the serological tests.

Q fever 261

3.17Q fever

(v) Skin testing and interpretation should only be carried out by experienced
personnel. For further information on training and accredited Q fever
immunisation service providers, contact your State or Territory Health
Department (see Appendix 1, Contact details for Australian, State and Territory
Government health authorities and communicable disease control). Skin testing is
performed by diluting 0.5mL of the Q-VAX Skin Test in 15mL of sodium
chloride (injection grade). Diluted Q-VAX Skin Test should be freshly prepared,
stored at +2C to +8C and used within 6 hours. 0.1mL of the diluted Q-VAX
Skin Test is injected intradermally into the volar surface of the forearm.
Commercial isopropyl alcohol skin wipes should not be used. If the skin is
not visibly clean, then methylated spirits may be used. A positive reaction is
indicated by any induration at the site of injection after 7days. Individuals
giving such a reaction must not be vaccinated, because they may develop severe
local reactions.

An indeterminate result, which occurs in only a small proportion of subjects, may


be the consequence of past infection with Q fever. It may also merely indicate the
presence in the subject of antibodies to antigens shared between C. burnetii and
other bacteria. Australian Q fever vaccine users have dealt with this finding in
one of two ways:
(a) Repeat the skin test and interpret as per the guidelines for initial testing.
Collect serum 2 to 3weeks later to look for a rise in titre of C. burnetii
antibodies in the IFA test, using Phase I and Phase II antigens, and
immunoglobulin class analysis. A significant increase (defined as a 4-fold rise
in titre of paired sera) indicates previous Q fever infection and vaccination is
then contraindicated.
(b) Vaccinate the subject using SC injection of a 5g (0.1mL) dose instead of
a 25g (0.5mL) dose of the vaccine. If there are no adverse effects (severe
local induration or severe systemic effects, perhaps accompanied by fever) 48
hours after the injection, a further 0.4mL (20g) dose of the vaccine is given
within the next 2 to 3weeks, ie. before the development of cell-mediated
immunity to the first dose.

Booster doses
Immunity produced by the vaccine appears to be long lasting (in excess of
5years). Until further information becomes available, revaccination or booster
doses of the vaccine are not recommended because of the risk of accentuated local
adverse events.

Contraindications
Q fever vaccine is contraindicated in the following:
individuals with a history of an illness suggestive of or proved to be Q fever,
those shown to be immune by either serological testing or sensitivity to the
organism by skin testing,
those who have been previously vaccinated against Q fever,
those with known hypersensitivity to egg proteins or any component of the
vaccine (Q-VAX may contain traces of egg protein, formalin, and sucrose).21
There is no information available on the accuracy of skin testing or the efficacy
and safety of Q fever vaccine use in individuals with impaired immunity. In
general, skin testing and Q fever vaccine should be avoided in such people.
The lower age limit for Q fever vaccine is not known. However, it is not
recommended for use in those aged <15years.

262 The Australian Immunisation Handbook 9th Edition

Precautions

Adverse events
Non-immune subjects very commonly show local tenderness (48%) and
erythema (33%) at the vaccination site. Local induration or oedema is uncommon
(<1%). General symptoms occur commonly in about 10% of vaccinees and may
include mild influenza-like symptoms such as headache (9%), fever (up to 2%),
chills and minor sweating.4,21
There are also 2 patterns of more significant adverse events among the estimated
more than 130000 individuals vaccinated from 19892004.5,17
The first and familiar pattern is the intensified local reaction at the injection
site which may occur shortly after inoculation in individuals sensitised
immunologically by previous infection or repeated vaccination. Rarely, an
immune abscess develops and requires excision and drainage. The acute
reactions may be accompanied by short-term systemic symptoms resembling the
post Q fever fatigue syndrome. Note, however, that not all those with positive
pre-vaccination skin and/or serological tests develop severe reactions. The
introduction of the pre-vaccination skin test at NIH/NIAID Rocky Mountain
Laboratory,30 later combined with antibody testing in Australia, has largely
eliminated reactions due to previous immune sensitisation. Despite this, the
adverse experience from the earlier American trials22 in which subjects were not
pre-tested, were vaccinated repeatedly or were inoculated with vaccines of a
different composition and larger bacterial mass, are still quoted in the general Q
fever literature as representative of a whole cell vaccine.
The second, much less frequent, pattern has been reported in people who were
skin and antibody test negative at the time of vaccination who did not have
any immediate reaction. Some 1 to 8months after vaccination, some vaccinees,
predominately women, developed an indurated lesion at the inoculation site.
At the time when the indurated lesion developed, the original skin test site
often became positive, presumably indicating a late developing cellular immune
response. These lesions were not fluctuant and did not progress to an abscess.
Most gradually declined in size and resolved over somemonths without
treatment. A few lesions were biopsied or excised and showed accumulations of
macrophages and lymphocytes.31,32

Use in pregnancy
Not recommended. Q fever vaccine contains inactivated products and inactivated
bacterial vaccines are not considered to be harmful in pregnancy. However, safety
of the vaccine in pregnancy has not been established. No information is available
on the use of Q fever vaccine during breastfeeding.

Q fever 263

3.17Q fever

Vaccination of subjects already immune to C. burnetii, as a result of either


previous infection or subjects being rendered hyperimmune by repeated
vaccination, may result in severe local or systemic adverse events.

Variations from product information


The product information for Q-VAX does not include the use of the reduced dose
of vaccine in individuals who have indeterminate results on either serological or
skin testing. However, this option has been used successfully by experienced Q
fever vaccinators.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

264 The Australian Immunisation Handbook 9th Edition

3.18 Rotavirus
Virology

Rotaviruses are shed in high concentrations in the stools of infected children and
are transmitted by the faecal-oral route, both through close person-to-person
contact and via fomites.6 Rotaviruses are probably also transmitted by other
modes, such as faecally contaminated food, water and respiratory droplets.7,8

Clinical features
Rotavirus is the predominant agent of severe dehydrating gastroenteritis in
infants and young children in both developed and developing countries.1,2
The spectrum of rotavirus illness ranges from asymptomatic infection, to mild,
watery diarrhoea of limited duration, to severe dehydrating diarrhoea with
vomiting, fever, electrolyte imbalance, shock and death. Rotavirus infections are
often more severe than other common causes of diarrhoea, and are more likely to
be associated with dehydration and hospitalisation.1,7 The incubation period is 1
to 3days, after which illness can begin abruptly with vomiting often preceding
the onset of diarrhoea.7 Up to one-third of patients have a temperature of >39C
in the first fewdays of illness. Symptoms generally resolve in 3 to 7days.

Epidemiology
Although individuals can be infected with rotavirus several times during their
lives, the first infection, typically between 3 and 36months of age, is most likely
to cause severe diarrhoea and dehydration.9,10 After a single natural infection,
40% of children are protected against any subsequent infection with rotavirus,
75% are protected against diarrhoea from a subsequent rotavirus infection, and
88% are protected against severe diarrhoea.10 Repeat infections provide even
greater protection. Disease is also less likely when reinfection occurs with a
serotype (G type) to which an individual has already been exposed.

Rotavirus 265

3.18Rotavirus

Rotaviruses are non-enveloped RNA viruses in the family Reoviridae.


Rotaviruses are classified according to the 2 surface proteins they contain:
VP7, the glycoprotein (G protein), and VP4, the protease-cleaved protein (P
protein). The G and P proteins are targets for neutralising antibodies thought
to be necessary for protection.1,2 Because the 2 gene segments that encode these
proteins can segregate independently, a typing system consisting of both P and
G types has been developed. Rotavirus strains are most commonly referred to
by their G serotype, with G1, G2, G3, G4 and G9 accounting for around 90% of
serotypes both globally and in Australia.3,4 The most common P types found
in combination with these G types are P1a[8] (found with all common G-types
except G2) or P1b[4], usually found in combination with G2.5

In Australia, the best available estimates are that approximately 10000


hospitalisations due to rotavirus in children <5years of age occur each year.11
As such, rotavirus accounts for around half the hospitalisations for acute
gastroenteritis of any cause in this age group.11,12 This translates to 3.8% of
children (1 in 27) being hospitalised with rotavirus gastroenteritis by the age
of 5years. In addition to hospitalised children, an estimated 115000 children
<5years of age visit a GP, and 22000 children require an Emergency Department
visit.11,13 On average, there is 1 death attributed to rotavirus each year in
Australia, but this is likely to be a minimum estimate.13
In temperate Australia, rotavirus infections follow a seasonal pattern, the peak
incidence being in mid to late winter. In the northern tropical and arid regions,
there is no consistent seasonal pattern and disease peaks are unpredictable.14
Epidemics of rotavirus gastroenteritis have occurred in Central Australia,
causing severe strain on healthcare services.15,16 Overall, Indigenous Australian
infants and children are hospitalised with rotavirus gastroenteritis about 3 to
5 times more commonly than their non-Indigenous peers, have a younger age
at hospitalisation, and longer duration of hospital stay (an average of 5days
compared with 2days for non-Indigenous infants).12,14,15,17
Children and adults with impaired immunity, such as those with congenital
immunodeficiency, or post haematopoietic or solid organ transplantation, are at
increased risk of severe, prolonged, and even fatal rotavirus gastroenteritis.1,18,19
Rotavirus is an important cause of nosocomial gastroenteritis,20-24 and can also
cause disease in adults, especially those caring for children, and outbreaks of
gastroenteritis in aged care facilities.1,25,26

Vaccines
Two oral rotavirus vaccines are available in Australia, and data on their
immunogenicity, safety and efficacy has been systematically reviewed.27 Both
vaccines are live attenuated vaccines administered orally to infants, but the
component vaccine viruses differ. Rotarix (GlaxoSmithKline) is a live attenuated
vaccine containing 1 strain of attenuated human rotavirus (G1P1[8] strain). The
human live attenuated strain protects against non G1 serotypes on the basis of
their common P[8] antigen and other epitopes involved in heterotypic immunity.
RotaTeq (CSL Biotherapies/Merck & Co Inc) is a pentavalent vaccine containing
5 human-bovine rotavirus reassortants with the human serotypes G1, G2, G3,
G4, and P1[8] and the bovine serotypes G6 and P7. The vaccine viruses replicate
in the intestinal mucosa and can be shed in the stool of vaccine recipients,
particularly after the first dose. Vaccine virus shedding is more common with
Rotarix and is detected in the stool aweek after vaccination in up to 80% of first
dose recipients, and in up to 30% of second dose recipients.27-29 RotaTeq is only
shed after the first dose (in up to 13% of recipients).30 There have been no studies
to assess the implications of shedding for horizontal spread to contacts.

266 The Australian Immunisation Handbook 9th Edition

Rotarix GlaxoSmithKline (live attenuated RIX4414 human rotavirus


strain expressing G1P1[8] outer capsid proteins). Each 1.0mL monodose
of the reconstituted vaccine contains not less than 106.0 CCID50 (cell culture
infectious dose 50%) of the RIX4414 strain; sucrose; dextran 40; sorbitol;
amino acids; Dulbeccos Modified Eagle Medium; calcium carbonate;
xanthan gum. Calcium carbonate buffer solvent (diluent) supplied for
reconstitution.
RotaTeq CSL Biotherapies/Merck & Co Inc (live, oral pentavalent
vaccine). Each 2.0mL monodose pre-filled dosing tube contains rotavirus
reassortants G1, G2, G3, G4 and P1[8] each with a minimum dose level of
at least 2.0 x 106 infectious units; sucrose; sodium citrate; sodium phosphate
monobasic monohydrate; sodium hydroxide; polysorbate 80; cell culture
media; trace amounts of fetal bovine serum. Also available in packs of 10
monodose pre-filled dosing tubes.

Transport, storage and handling


Transport both vaccines according to National Vaccine Storage Guidelines: Strive for
5.34 Store at +2C to +8C. Do not freeze. Protect from light.

Rotavirus 267

3.18Rotavirus

Current oral rotavirus vaccines are underpinned by decades of developmental


work.31 Randomised placebo-controlled studies of both vaccines have
documented their efficacy and safety in the prevention of gastroenteritis caused
by rotavirus.27,30,32 A vaccination course prevents rotavirus gastroenteritis of any
severity in approximately 70% of recipients over the following 1 to 2years. The
efficacy against severe rotavirus gastroenteritis and against hospitalisation for
rotavirus gastroenteritis is higher, ranging from 85 to 100% in clinical trials in
many different countries.27,30,32,33 Efficacy in the prevention of hospitalisation from
rotavirus gastroenteritis ranged from 85 to 100%.27,30,32,33 Vaccination was also
highly effective in preventing Emergency Department and clinic/GP visits.30,33
Overall, rotavirus vaccination prevented around half (4258%) of hospital
admissions for acute gastroenteritis of any cause in young children, suggesting
that rotavirus is responsible for more gastroenteritis than detected using routine
testing and admission practices.30,32,33 In randomised control trials, a degree of
protection against rotavirus gastroenteritis was also observed in infants who
received fewer than the recommended number of doses of rotavirus vaccines.
In the available clinical trials, no statistically significant differences were found
between the 2 vaccines with regard to protective efficacy by serotype.27,30,32
The efficacy and safety of both rotavirus vaccines have been evaluated only
in clinical trials in which infants received vaccine within specified age limits.
There are no data on the use of rotavirus vaccines outside these age ranges (see
Recommendations and Adverse events below).

Dosage and administration


Rotavirus vaccines are for oral administration only. Under no circumstances
should rotavirus vaccines be injected.
There are no restrictions on the infants consumption of food or liquid, including
breast milk, either before or after vaccination with either rotavirus vaccine.7,35
Rotarix is recommended for use in a 2-dose course (at 2 and 4months of age).
It is presented as a white powder for reconstitution with a separately supplied
diluent, and a transfer adapter. The syringe/oral plunger containing the diluent
is attached to the vial of lyophilised powder via the transfer adapter, and
following reconstitution the 1mL dose of vaccine should be administered orally
via the syringe/oral plunger onto the inside of the infants cheek.
RotaTeq is recommended for use in a 3-dose course (at 2, 4, and 6months of age).
It is supplied in a container consisting of a squeezable plastic, latex-free dosing
tube with a twist-off cap, allowing for direct oral administration of the 2mL dose
onto the inside of the infants cheek. RotaTeq does not require reconstitution or
dilution. RotaTeq is a pale yellow, clear liquid that may have a pink tint.
Rotavirus vaccines can be co-administered with other vaccines included on the
NIP schedule at 2 and 4months of age (Rotarix) or 2, 4 and 6months of age
(RotaTeq). The available evidence from clinical trials suggests co-administration
of oral rotavirus vaccines is safe and effective and does not interfere with the
immune response to the other vaccine antigens (DTPa, Hib, IPV, hepB, and
7vPCV).28,29,35

Recommendations
(i) Routine infant vaccination (Safety-Grade B)(EfficacyGrade B)(Immunogenicity-not assessed)27
Administration of a course of oral rotavirus vaccination is recommended for all
infants in the first half of the first year of life. Vaccination of older infants and
children is not recommended as there are theoretical concerns regarding use in
older age groups (see Adverse events below). Vaccination should occur at either
2 and 4months of age (Rotarix), or 2, 4 and 6months of age (RotaTeq), according
to the following schedules (see also Table 3.18.1):
Rotarix (human monovalent rotavirus vaccine)
The vaccination course of Rotarix consists of 2 doses at approximately 2 and
4months of age. The first dose should be given between 6 and 14weeks of
age, and the second dose should be given by the end of the 24thweek of age
(6months). The interval between the 2 doses should not be less than 4weeks.
RotaTeq (pentavalent human-bovine reassortant rotavirus vaccine)
The vaccination course of RotaTeq consists of 3 doses at approximately 2, 4, and
6months of age. The first dose should be given between 6 and 12weeks of age,

268 The Australian Immunisation Handbook 9th Edition

and all doses should be given by the end of the 32ndweek of age (~7.5months).
The interval between doses should be 4 to 10weeks.
Table 3.18.1: Age limits for dosing of oral rotavirus vaccines
Doses

Age of
routine oral
administration

Age limits for dosing


1st dose

2nd dose

3rd dose

Minimum
interval
between
doses

Rotarix
(GlaxoSmithKline)

2 oral
doses
(1mL/
dose)

2 and
4months

614*weeks

1024*weeks

None

4weeks

RotaTeq
(CSL Biotherapies/
Merck & Co Inc)

3 oral
doses
(2mL/
dose)

2, 4 and
6months

612weeks

1032weeks

1432weeks

4weeks

The upper age limit for receipt of the first dose of RotaTeq is 12.9weeks, that is up to the
anniversary of the 13thweek of age. The second dose of vaccine should preferably be given
by 28weeks of age to allow for a minimum interval of 4weeks before receipt of the third
dose, and the upper age limit for either the second or third doses is 32.9weeks, that is by the
anniversary of the 33rdweek of age.

For infants in whom the first dose of rotavirus vaccine is inadvertently


administered at an age greater than the suggested cut-off (14weeks for Rotarix
or 12weeks for RotaTeq), the remaining vaccine doses should be administered
as per the schedule, providing the minimum interval between doses can be
maintained, and the course completed within the recommended age limits. The
timing of the first dose should not affect the safety and efficacy of the second and
third dose.7 Infants who develop rotavirus gastroenteritis before receiving the
full course of rotavirus vaccinations should still complete the full 2- or 3-dose
schedule (dependent on the brand of vaccine) because one rotavirus infection
only provides partial immunity.7

(ii) Catch-up (no studies)


Routine catch-up or primary vaccination of older children is not recommended.
Infants should commence the course of rotavirus vaccination within the
recommended age limits for the first dose. It is also necessary to ensure that
doses are not given beyond the upper age limits for the final dose of the vaccine
course (see (i) above). This is based on theoretical concerns regarding possible
adverse events in older age groups (see Adverse events below), and because
the safety of rotavirus vaccination in older infants and children has not been
established.

Rotavirus 269

3.18Rotavirus

* The upper age limit for receipt of the first dose of Rotarix is 14.9weeks, that is up to the
anniversary of the 15thweek of age and the upper age limit for receipt of the second dose of
Rotarix is 24.9weeks, that is up to the anniversary of the 25thweek of age.

(iii) Premature infants (Safety-Grade C)


(Efficacy-Grade C)(Immunogenicity-not assessed)27
Vaccination of preterm infants using either available rotavirus vaccine is
indicated at a chronologic age of at least 6weeks if clinically stable. Premature
infants (<37weeks gestation) appear to be at increased risk of hospitalisation
from viral gastroenteritis.36 In clinical trials, RotaTeq or placebo was administered
to 2070 preterm infants (2536weeks gestational age; median 34weeks) who
experienced rates of adverse events after vaccination similar to matched placebo
recipients.7,27 Efficacy against rotavirus gastroenteritis of any severity was
evaluated in only a small subset of premature infants and appeared comparable
to efficacy in term infants (70%; 95% CI: -15%95%). These conclusions would
also be expected to apply to Rotarix vaccine. If standard infection control
precautions are maintained, administration of rotavirus vaccine to hospitalised
infants, including hospitalised premature infants, would be expected to carry a
low risk for transmission of vaccine viruses (see Precautions below).

Contraindications
The only absolute contraindications to rotavirus vaccines are:
anaphylaxis following a previous dose of either rotavirus vaccine, or
anaphylaxis following any vaccine component.

Precautions
(i) Acute gastroenteritis
Infants with moderate to severe acute gastroenteritis should not be vaccinated
until after recovery from their acute illness. Infants with mild gastroenteritis
(including mild diarrhoea) can be vaccinated. The use of rotavirus vaccines has
not been studied in infants with acute gastroenteritis.

(ii) Moderate to severe illness


As with other vaccines, infants with a moderate to severe illness should be
vaccinated after recovery. In addition to the factors mentioned above in (i), this
avoids superimposing potential adverse events related to vaccination with the
concurrent illness.

(iii) Underlying conditions predisposing to severe rotavirus gastroenteritis


Conditions predisposing to severe or complicated rotavirus gastroenteritis
include metabolic disorders or chronic gastrointestinal disease, such as
Hirschsprungs disease, malabsorption syndromes or short gut syndrome.1
Although the safety and efficacy of rotavirus vaccines have not been studied in
such infants, because they are at greater risk of serious rotavirus disease over an
extended age range, the potential benefits of vaccination at an age older than the
upper limits recommended in Table 3.18.1 are likely to be substantial. Vaccination
of such children at an older age may be judged by clinicians to warrant

270 The Australian Immunisation Handbook 9th Edition

discussion with parents on a case by case basis (see Variations from product
information below).

(iv) Infants with impaired immunity


There are no studies of the safety or efficacy of the currently available rotavirus
vaccines in infants with impaired immunity. As with other live viral vaccines,
there are theoretical concerns that vaccine virus-associated gastrointestinal
disease could occur in infants with severely impaired immunity who receive
rotavirus vaccines. However, the theoretical risk for vaccine virus-associated
disease in immune-impaired vaccinated infants is likely to be less than their
risk from being exposed to disease from natural infection. Risks and benefits of
vaccination should be considered in the context of the infants specific immune
impairment with appropriate specialist advice7 (see (v) below, and Section 2.3.3,
Vaccination of individuals with impaired immunity due to disease or treatment).
Infants living in households with people who have impaired immunity should
be vaccinated. In general, household members with impaired immunity are
afforded protection by vaccination of young children in the household. This
outweighs the small risk for transmitting vaccine virus shed in stool to the
household member with impaired immunity. The theoretical risk for vaccine
virus-associated disease in contacts with impaired immunity is considered less
than their risk of being exposed to disease from natural infection. However, there
have been no studies to specifically address this question.7 (See also Section 2.3.3,
Vaccination of individuals with impaired immunity due to disease or treatment.)

(vi) Recent administration of antibody-containing blood products


Infants who have recently received antibody-containing blood products and are
at an eligible age should be vaccinated. The interval between vaccination and
receipt of the blood product should be as long as possible, but without delaying
administration of vaccine beyond the suggested age limits for dosing (as per
Table 3.18.1 above). This recommendation for maximising the interval is based on
theoretical concern that passively acquired antibody to rotavirus may interfere
with vaccine immunogenicity.7

(vii) Hospitalised infants


If a recently vaccinated child is hospitalised for any reason, no precautions other
than routine standard precautions need be taken to prevent the spread of vaccine
virus in the hospital setting. Administration of rotavirus vaccine to hospitalised
infants, including hospitalised premature infants, is likely to carry a low risk
for transmission of vaccine viruses if standard infection control precautions are
maintained (see Vaccines above).

Rotavirus 271

3.18Rotavirus

(v) Infants living in households with people with impaired immunity

(viii) Exposure of pregnant women to vaccinated infants


Infants living in households of pregnant women can receive rotavirus vaccines.
Most pregnant women will have pre-existing immunity to rotavirus but
avoidance of wild-type infection through the vaccination of infant contacts
may benefit adults, including pregnant women, and outweighs any theoretical
concern regarding exposure to vaccine viruses.

(ix) Regurgitation of vaccine dose


Readministration of the vaccine is not necessary after regurgitation, spitting out,
or vomiting of a rotavirus vaccine. This is because there are limited data available
on the safety of administering higher than the recommended dose of rotavirus
vaccines. There are no studies of the efficacy of a partially administered dose(s).

Adverse events
(i) Intussusception (IS)
Current evidence indicates that intussusception (IS, a form of bowel obstruction)
is not associated with either Rotarix or RotaTeq vaccines, especially when given
to infants within the age limits studied in clinical trials.27,30,32 Post-licensure data
in larger numbers of children will monitor if there is an increased risk of IS
following rotavirus vaccination, particularly among those inadvertently receiving
doses outside the recommended age limits. Concern about association between
IS and rotavirus vaccines arose because a tetravalent rhesus-reassortant vaccine,
called RotaShield, licensed in the United States (but not elsewhere) in 199899,
was associated with IS in approximately 1 in 10000 vaccine recipients.37 The
greatest risk of IS occurred within 3 to 14days after the first dose, with a smaller
risk after the second dose.37,38
There is evidence that when the first dose of RotaShield was given at >3months
of age, the risk of intussusception was increased.38 The pathogenesis of
RotaShield-associated intussusception has not been determined. However,
the current rotavirus vaccines (RotaTeq and Rotarix) differ in composition to
RotaShield, which was also more reactogenic.39-41 The large-scale safety studies
of the 2 current rotavirus vaccines included approximately 140000 infants,
and found the risk of IS in vaccine recipients to be similar to that of placebo
recipients, and less than that estimated for RotaShield.27,30,32 To minimise
background rates of IS, the clinical trials of Rotarix and RotaTeq limited
administration of the first dose of vaccine to infants under 14 and 12weeks
of age, respectively, and did not give subsequent doses to infants beyond a
certain age (24weeks for Rotarix and 32weeks for RotaTeq).27,30,32 As such,
data on safety of these vaccines in older infants is not currently available (see
Recommendations above).

272 The Australian Immunisation Handbook 9th Edition

(ii) Other adverse events


Vaccine recipients developed gastrointestinal symptoms such as diarrhoea or
vomiting in theweek after rotavirus vaccination more commonly than placebo
recipients (increased risk of up to 3%).27,30,32 Fever was not significantly more
common in rotavirus vaccine recipients compared with placebo recipients in
clinical trials of both available vaccines.27,30,32

Interchangeability of rotavirus vaccines

Variations from product information


The product information for Rotarix states that the vaccine should not be
administered to subjects with chronic gastrointestinal disease. NHMRC
recommends that pre-existing chronic gastrointestinal disease is not considered
to be a contraindication to rotavirus vaccination (see Precautions above).

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

Rotavirus 273

3.18Rotavirus

Completion of a course of rotavirus vaccine should be with vaccine from the


same manufacturer whenever possible. There are no studies that address the
interchangeability of the 2 available rotavirus vaccines. However, if either dose 1
or 2 of vaccine is given as RotaTeq, a third dose of either rotavirus vaccine should
be given, provided that the upper age limit and inter-vaccine interval, as defined
above in Recommendations, Table 3.18.1, are met.

3.19 Rubella
Virology
Rubella is an enveloped togavirus, genus Rubivirus. The virus has an RNA
genome and is closely related to group A arboviruses, but does not require
a vector for transmission. It is relatively unstable, and is inactivated by lipid
solvents, trypsin, formalin, extremes of heat and pH, amantadine and UV light.1

Clinical features
Rubella is generally a mild and self-limiting infectious disease.2 It causes a
transient, generalised, erythematous, maculopapular rash, lymphadenopathy
involving the post-auricular and sub-occipital glands, and, occasionally,
arthritis and arthralgia. Other complications, such as neurological disorders and
thrombocytopenia, may occur but are rare. Clinical diagnosis is unreliable since
the symptoms are often fleeting and can be caused by other viruses; in particular,
the rash is not unique to rubella and may be absent.1,2 Up to 50% of rubella virus
infections are subclinical or asymptomatic.1 A history of rubella should, therefore,
not be accepted without serological evidence of previous infection.1 The
incubation period is 14 to 21days, and the period of infectivity is from 1week
before until 4days after the onset of the rash.2 Rubella infection in pregnancy can
result in fetal infection resulting in congenital rubella syndrome (CRS) in a high
proportion of cases (see Rubella infection in pregnancy below).

Epidemiology
Rubella occurs worldwide and is spread from person to person by droplet
contact and possibly air-borne transmission of infectious respiratory secretions.1
In temperate climates, the incidence is highest in late winter and early spring.3
The incidence of rubella has fallen rapidly since vaccine licensure, and there
has been a shift in the age distribution of cases, with comparatively more cases
being seen in older age groups, particularly the 2024 year age group.4 In the
early 1990s, rubella epidemics were reported in those States where rubella
was notifiable.5 Over 3000 cases per year were reported between 1992 and
1995.5 In 20042005, rubella notifications were the lowest yet recorded with 31
confirmed cases being reported in each year (0.15 per 100000 per year).4 This
low notification rate most likely reflects the high vaccine coverage achieved and
sustained with the National Measles Control Campaign in late 1998.3,6,7
The number of cases of congenital rubella syndrome has also fallen rapidly
since rubella vaccine licensure in Australia. Successful vaccination campaigns
and high vaccination coverage resulted in no cases of congenital rubella
syndrome occurring in infants of Australian-born mothers between 1998 and
2002. However, 5 cases resulting from infection acquired outside of Australia

274 The Australian Immunisation Handbook 9th Edition

were reported during this time.8,9 Between 2003 and 2005, an additional 5 cases
were reported from infection that occurred in Australia9-11 which reinforces the
need for high vaccination coverage of women of child-bearing age (see Rubella
infection in pregnancy below).
The rubella virus was isolated in cell culture in 1962, and vaccines prepared
from strains of attenuated virus have been approved for use in Australia since
1970. Mass vaccination of schoolgirls commenced in 1971.1,12 Non-pregnant,
seronegative adult women were also vaccinated. These programs were
successful and there was a significant reduction in the incidence of congenital
rubella syndrome from 1977.13-15 There has also been a significant increase in the
percentage of pregnant women immune to rubella (in NSW from 82% in 1971
to 96% in 1983). Based on a recent study in Melbourne, it was estimated that, in
2000, only 2.5% of all women in Australia of child-bearing age were seronegative.
However, susceptibility was higher among overseas-born women, and has been
reported as higher among some Indigenous women.16,17

Rubella infection in pregnancy


Maternal rubella infection in the first 8 to 10weeks of pregnancy results in
fetal damage in up to 90% of affected pregnancies, and multiple defects are
common.19-21 The risk of damage declines to 10 to 20% by 16weeks gestation.
After this stage of pregnancy, fetal damage is rare but has been reported up to
20weeks gestation.19 The characteristics of congenital rubella syndrome include
intellectual disabilities, cataracts, deafness, cardiac abnormalities, intrauterine
growth retardation and inflammatory lesions of the brain, liver, lungs and
bone marrow.19 Any combination of these defects may occur, but defects which
commonly occur alone following infection after the first 8weeks of pregnancy
are perceptive deafness and pigmentary retinopathy. Some infected infants may
appear normal at birth, but defects, especially sensorineural deafness, may be
detected later.22
Rubella reinfection can occur in individuals who have both natural and vaccineinduced antibody.19 Occasional cases of congenital rubella syndrome after
reinfection in pregnancy have been reported. However, fetal damage is very rare in
cases of infection in women in whom antibody has previously been detected.20,23-25

Rubella 275

3.19Rubella

Many adolescent and young adult males are not immune to rubella because
they did not receive an MMR vaccine.18 The MMR vaccination program for all
adolescents replaced the rubella program for girls in 1993/94.12 A serosurvey
conducted in 1999 showed that only 84% of males aged 1418years (compared to
95% of females) and 89% of males aged 1949years (compared to 98% of females)
were immune to rubella.18 For this reason, adolescent and young adult males,
as well as females, who do not have a documented history of receipt of 2 doses
of MMR, should receive MMR vaccine (see Recommendations below). This is
both for their own protection and to prevent transmission of the infection in the
community (see The public health management of rubella below).

All pregnant women with suspected rubella or exposure to rubella should be


serologically tested, irrespective of a history of previous vaccination, clinical
rubella or a previous positive rubella antibody result (see Serological testing
for rubella below). This is because the rash of rubella is not diagnostic,
asymptomatic infection can occur, and acute rubella can be confirmed only by
laboratory tests.19,23,24 Pregnant women should be counselled to restrict contact
with individuals with confirmed, probable or suspected rubella for 6weeks (2
incubation periods).26 Counselling of pregnant women with confirmed rubella
regarding the risk to the fetus should be given in conjunction with the womans
obstetric service.

Serological testing for rubella


A number of commercial assays for testing immunity to rubella are available.
These vary according to the method used to determine the positive cut-off value
(the WHO cut-off is 10 IU/mL but, at present, there is no recommended Australian
minimal level). Available data support the presumption that an antibody level
found by use of a licensed assay to be above the standard positive cut-off for that
assay can be considered evidence of past exposure to rubella virus.23 Antibody
levels below the cut-off are likely not to be protective, particularly if the antibodies
have been generated by vaccination rather than by natural infection, and MMR
vaccine (or MMRV if protection against varicella is required in children 12months
to 12years of age) should be administered according to the Recommendations
below. Expert consultation and referral of sera to a reference laboratory are
recommended if there is a difficulty interpreting results.
Acute rubella infection is indicated by presence of rubella IgM or 4-fold or
greater increase in rubella IgG. Rubella IgM may not appear until aweek after
clinical symptoms. Sera for IgG testing should be taken 7 to 10days after onset of
illness and repeated 2 to 3weeks later. The most recent date of potential exposure
should be obtained, if possible, to calculate the potential incubation period. As
some patients may have more than 1 exposure to a person with a rubella-like
illness, and because exposure may occur over a prolonged period, it is important
to ascertain the dates of the first and last exposures.26
Seronegative women of child-bearing age should be vaccinated (see
Recommendations below) and tested for seroconversion 8weeks after
vaccination. All women should be informed in writing of the result of their
antibody test. Women should be screened for rubella antibodies shortly before
every pregnancy, or early in the pregnancy, or if pregnancy is contemplated,
irrespective of a previous positive rubella antibody result.15,19 Very occasionally,
errors may result in patients who are seronegative being reported as seropositive.
Where possible, specimens from pregnant women should be stored until the
completion of the pregnancy.
Serological testing of pregnant women exposed to rubella should always
be performed (see Rubella infection in pregnancy above). A blood sample

276 The Australian Immunisation Handbook 9th Edition

should be taken and sent to the laboratory with the date of the last menstrual
period and the date of presumed exposure (or date of onset of symptoms).26
If the woman has an antibody titre below the protective level, or a low level
of antibodies and remains asymptomatic, a second blood specimen should
be collected 28days after the exposure (or onset of symptoms) and tested in
parallel with the first. If the woman develops symptoms, the specimen should
be collected and tested as soon as possible. A third blood specimen may be
required in some circumstances.24

Vaccines
Rubella vaccine is available as either MMR vaccine or as a monovalent rubella
vaccine. It is anticipated that combination measles-mumps-rubella-varicella
(MMRV) vaccines will become available in the near future. A single dose of
rubella vaccine produces an antibody response in more than 95% of vaccinees,
but antibody levels are lower than after natural infection.19,23,24 Vaccine-induced
antibodies have been shown to persist for at least 16years in the absence of
endemic disease.23,24,27,28 Protection against clinical rubella appears to be longterm in those who seroconvert.19
Monovalent rubella vaccine

Combination measles-mumps-rubella vaccine


Priorix (MMR) GlaxoSmithKline (live attenuated measles virus (Schwarz
strain), RIT 4385 strain of mumps virus (derived from the Jeryl Lynn strain)
and the Wistar RA 27/3 rubella virus strain). Each 0.5mL monodose of the
reconstituted, lyophilised vaccine contains not less than 103.0 CCID50 (cell
culture infectious dose 50%) of the Schwarz measles, not less than 103.7
CCID50 of the RIT 4385 mumps and not less than 103.0 CCID50 of the Wistar
RA 27/3 rubella virus strains; lactose; neomycin; amino acids; sorbitol and
mannitol as stabilisers.

Transport, storage and handling


Transport both vaccines according to National Vaccine Storage Guidelines: Strive for
5.29 Store at +2C to +8C. Protect from light. Do not freeze. Reconstituted vaccine
should be used immediately, but can be stored at +2C to +8C for up to 8 hours
before use.

Rubella 277

3.19Rubella

Meruvax II CSL Biotherapies/Merck & Co Inc (rubella virus vaccine).


Each 0.5mL monodose of the reconstituted, lyophilised vaccine contains
not less than 1000 TCID50 (tissue culture infectious dose 50%) of attenuated
rubella virus (Wistar RA 27/3 strain); 25g neomycin; 3mg human serum
albumin; sorbitol and gelatin as stabilisers.

Dosage and administration


For both children and adults, the dose of MMR and monovalent rubella vaccine
is 0.5mL, administered by either SC or IM injection.
MMR and monovalent rubella vaccine can be given at the same time as other
vaccines (including DTPa, hepatitis B, MenCCV and varicella), using separate
syringes and injection sites. If MMR or monovalent rubella vaccine is not given
simultaneously with other live viral parenteral vaccines (eg. varicella vaccine),
they should be given at least 4weeks apart (see Precautions below).

Recommendations
The principal aim of rubella vaccination is to prevent congenital rubella
syndrome by stopping the circulation of rubella virus in the community.
Susceptible pregnant women will continue to be at risk of rubella infection in
pregnancy until the transmission of rubella virus is interrupted by a sufficiently
high uptake of rubella-containing vaccine in children and adults of both sexes.

(i) Routine vaccination of children


Two doses of rubella-containing vaccine are recommended for all children. The
first dose should be given at 12months of age and the second dose at 18months
of age (MMR or MMRV when available). The minimum interval between the
first and second doses of MMR or MMRV is 4weeks. A history of rubella is not a
contraindication to vaccination. Individuals who are already immune to rubella
have no increased risk of side effects from vaccination.19,23

(ii) Vaccination of women of child-bearing age


Every effort should be made to identify non-pregnant seronegative women of
child-bearing age. The following women are more likely to be seronegative to
rubella: women born overseas (especially in Asia, Pacific islands, sub-Saharan
Africa and South America) who have entered Australia after the age of routine
vaccination; non-English speaking women; women over the age of 35; and
Muslim women.5,13,14,16,30 Seronegative women should be given MMR vaccine
and advised not to become pregnant for 28days after vaccination. Monovalent
rubella vaccine can be used where there is a contraindication to the measles
or mumps components of MMR. Vaccinated women should be tested for
seroconversion 6 to 8weeks after vaccination (see Serological testing for rubella
above). Women who have negative or very low antibody levels after vaccination
should be revaccinated. If their antibody levels remain low after a second
vaccination, it is unlikely that further vaccinations will improve this.19 Although
2 doses of MMR vaccine are routinely recommended, if rubella immunity
is demonstrated after receipt of 1 dose of a rubella-containing vaccine, no
further dose is required, unless indicated by subsequent serological testing (see
Serological testing for rubella above).

278 The Australian Immunisation Handbook 9th Edition

(iii) Vaccination of adolescent and adult males


All males born during or after 1966 require 2 doses of MMR at least 4weeks
apart if they have no record of receiving the vaccine, as they are especially likely
to be non-immune to rubella (see Epidemiology above).

(iv) Vaccination post-partum


Women found to be seronegative on antenatal rubella immunity testing should
be vaccinated after delivery and before discharge from the maternity unit.
MMR vaccine is recommended, although monovalent rubella vaccine can also
be used for this purpose. These women should be tested for rubella immunity
6 to 8weeks after vaccination (see Vaccination of women of child-bearing age
above). Anti-D immunoglobulin does not interfere with the antibody response
to vaccine.1,19 If anti-D immunoglobulin is also required, the two may be given at
the same time in different sites with separate syringes, or at any time in relation
to each other24 (see Contraindications below).

(v) Vaccination of healthcare workers and people working with children

For further recommendations related to MMR vaccination, see Chapter 3.11,


Measles.

Contraindications
Vaccination is contraindicated in the following circumstances:

(i) Allergy to vaccine components


anaphylaxis following a previous dose of rubella, MMR or MMRV, or
anaphylaxis following any vaccine component.

(ii) People with impaired immunity


Rubella-containing vaccine should not be administered to patients with
congenital or acquired impaired immunity (see Section 2.3.3, Vaccination of
individuals with impaired immunity due to disease or treatment). This includes
those receiving high-dose corticosteroid or immunosuppressive treatment,
general radiation, malignant conditions of the reticuloendothelial system (such
as lymphoma, leukaemia, Hodgkins disease), or in cases where the normal

Rubella 279

3.19Rubella

All healthcare staff and people working with children, born during or since 1966,
including medical, nursing, and other health professional students, either without
vaccination records or seronegative upon screening, should receive 2 doses of
MMR vaccine, both for their own protection and to avoid the risk of transmitting
rubella to pregnant patients and/or colleagues31 (see Table 2.3.6 Recommended
vaccinations for those at risk of occupationally acquired vaccine-preventable diseases).
Preferably, MMR should be used. Where necessary, those vaccinated can be tested
for seroconversion 8weeks after vaccination and revaccinated if seronegative (see
Vaccination of women of child-bearing age above).

immunological mechanism may be impaired, as in hypogammaglobulinaemia.1,23


Rubella vaccine or MMR may be given to HIV-positive individuals unless they
have severely impaired immunity.23 (For further information on MMR and
MMRV vaccines, see Chapter 3.11, Measles and Chapter 3.24, Varicella).

(iii) Recent administration of antibody-containing blood product


Rubella-containing vaccine should not be given within at least 3months after an
injection of immunoglobulin, other antibody-containing blood product, or wholeblood transfusion, because the expected immune response may be impaired.19,24
The recommended intervals for receipt of rubella-containing vaccines after receipt
of blood products are given in Table 2.3.5 Recommended intervals between either
immunoglobulins or blood products and MMR, MMRV or varicella vaccination. Rubellacontaining vaccines may be administered concomitantly with, or at any time in
relation to, anti-D immunoglobulin, but at a separate injection site. However,
women who have received anti-D immunoglobulin should be serologically tested
8weeks after vaccination to ensure that seroconversion has occurred.1,23

(iv) Pregnant women


MMR and monovalent rubella vaccines should not be given to a woman known
to be pregnant, and pregnancy should be avoided for 28days after vaccination
(see Use in pregnancy below).1,32 Data on the use of MMRV vaccines in
individuals >12years of age are not available.

Precautions
If MMR or monovalent rubella vaccine is not given simultaneously with
other live viral parenteral vaccines (eg. varicella vaccine), they should be
given at least 4weeks apart.
Breastfeeding is not a contraindication to rubella vaccination. The rubella
vaccine virus may be secreted in human breast milk, and there have been
rare cases of transmission of vaccine virus through breast milk reported.
However, these infections have been mild.1
There is no risk to pregnant women from contact with recently vaccinated
individuals. The vaccine virus is not transmitted from vaccinees to
susceptible contacts.1
For precautions related to MMR and MMRV vaccines, see Chapter 3.11, Measles
and Chapter 3.24, Varicella.

Adverse events
Mild adverse events such as fever, sore throat, lymphadenopathy, rash, arthralgia
and arthritis may occur after vaccination.1,23 Symptoms most often begin 1 to
3weeks after vaccination and are usually transient. Joint symptoms are more
common in adults, especially women (10 to 25%, very common) than in children
(0.3%, uncommon).1,23 Thrombocytopenia, that is usually self limiting, has been

280 The Australian Immunisation Handbook 9th Edition

reported rarely after rubella vaccine.23 Very rarely, neurological symptoms have
been reported, but a causal relationship has not been established.23
For adverse events related to MMR and MMRV vaccines, see Chapter 3.11,
Measles and Chapter 3.24, Varicella.

The public health management of rubella


All cases of suspected rubella infection should be laboratory tested and false
positive results excluded. Infected individuals should be excluded from school/
work/institution and should avoid contact with women of child-bearing age for
at least 4days after the onset of the rash.26
All contacts should be identified, especially those who are pregnant. If a contact
is pregnant, see Rubella infection in pregnancy above. All contacts >12months
of age without adequate proof of immunity should receive 1 dose of MMR (or
MMRV, when available, in those 12months to 12years of age). This will not
prevent rubella disease if already exposed. If vaccination is refused, the contact
should avoid further contact with cases until at least 4days after onset of the rash
in the case.
Exposed healthcare workers without adequate proof of immunity should be
excluded from work for 21days from exposure or for at least 4days after the
onset of a rash.26

Use in pregnancy

Use of normal human immunoglobulin


(NHIG) to prevent rubella
Post-exposure prophylaxis with NHIG does not prevent infection in non-immune
contacts and is, therefore, of little value for protection of pregnant women
exposed to rubella.23 It may, however, prolong the incubation period, which
may marginally reduce the risk to the fetus. It may also reduce the likelihood
of clinical symptoms in the mother. NHIG should only be used if termination

Rubella 281

3.19Rubella

Vaccination should be avoided in early pregnancy.1 However, active


surveillance in the USA, UK and Germany indicates that no case of vaccineinduced congenital rubella syndrome occurred among more than 500 women
inadvertently vaccinated with rubella vaccine during pregnancy, whose
pregnancies continued.33 In a recent Iranian study performed after mass
vaccination with a measles-rubella vaccine, 117 susceptible women were
inadvertently vaccinated while pregnant or became pregnant 3months after
vaccination. There were no CRS-related abnormalities among the infants born
to these women.34 Based on this evidence, the vaccine cannot be considered to
be teratogenic, and termination of pregnancy following inadvertent vaccination
is not indicated1,24 (see Section 2.3.2, Vaccination of women planning pregnancy,
pregnant or breastfeeding women, and preterm infants).

for confirmed rubella would be unacceptable under any circumstances. In such


cases, IM administration of 20mL of NHIG within 72 hours of rubella exposure
might reduce but will not eliminate the risk for rubella.23 Serological followup of recipients is essential, and should continue for up to 2months.
There is some evidence to suggest that, in outbreak situations, pre-exposure
NHIG may be effective in preventing infection in women who are likely to be
pregnant, and its use may be indicated for such women with low antibody titres
in high-risk occupations.35

Variations from product information


The product information recommends that women of child-bearing age should
be advised not to become pregnant for 3months after vaccination with rubella,
MMR or MMRV vaccines, whereas NHMRC recommends 28days.32
The product information for Meruvax II recommends the vaccine be given by SC
injection, but NHMRC recommends administration by either SC or IM injection.
The product information for Meruvax II states that there is no reason to
revaccinate individuals who were vaccinated originally when 12months of age
or older. However, NHMRC recommends routine administration of a second
dose of rubella vaccine when given as MMR or MMRV to children.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

282 The Australian Immunisation Handbook 9th Edition

3.20Smallpox
Virology
The smallpox or variola virus is one of the poxviruses, a group characterised by
large brick-shaped virus particles, which includes the agents vaccinia, monkeypox,
mousepox and cowpox. The virus is inhaled into the respiratory tract, multiplies
in local lymph nodes and then seeds to the reticuloendothelial system. During the
clinical prodrome the virus then circulates to the skin and mucous membranes
where the cell destruction produces the characteristic vesicular lesions.1

Clinical features
An incubation period of about 12days is abruptly followed by the prodrome, a
2 to 5 day period of high fever, malaise and severe headache. Then follows the
pharyngeal enanthum and, a day later, the skin rash begins as small red macules
before progressing to papules, vesicles and finally pustules over the next 4 to
7days.1 Death follows in about 20% of cases. The diagnosis is made by collecting
vesicular fluid for examination by electron microscopy, or for detection of viral
nucleic acids by amplification techniques. Diseases that are most likely to mimic
smallpox in Australian populations are varicella and drug eruptions.

Epidemiology
Smallpox, a disease only of humans, was declared eradicated in 1979 after an
intense international campaign of detection and vaccination. The disease would
now be of only historical interest if not for concerns that illicit laboratory stocks
of the virus may exist and may be used as biological weapons.2 In thedays of
endemic disease in rural areas, each case of smallpox would generate several
more cases among family and friends attending the victim, who was usually
bed-bound from the onset of the prodromal illness. The epidemiology of
disease spread by bioterrorists may be quite different. Patients hospitalised
in the prodromal period may widely transmit the virus during coughing, as
demonstrated in an outbreak in a German hospital after admission of one patient
with unrecognised smallpox.3

Little is known of the origin of vaccinia virus, the poxvirus used to immunise
humans against smallpox. Despite its name, which has been given generally to
compounds (vaccines) which induce artificial immunity, it is not cowpox.4-6
Australia has stocks of smallpox vaccine for use in an emergency situation only.4-6
The USA smallpox vaccine Dryvax, has been used to vaccinate Australian laboratory
personnel working with poxviruses. This vaccine contains vaccinia which produces
cross-immunity against variola. Dryvax is a freeze-dried formulation.

Smallpox 283

3.20 Smallpox

Vaccines

The duration of immunity is uncertain. A recent review, examining the frequency


of adverse events after vaccination with different vaccinia strains, reported that
the Lister strain vaccine is associated with a higher risk of severe adverse events,
in particular postvaccinal encephalopathy.7

Transport, storage and handling


Transport according to Guidelines for smallpox outbreak, preparedness, response and
management.4 Smallpox vaccine should be kept frozen at 30C. The shelf life of
the vaccine is 24 hours once thawed and, once thawed, the vaccine should be
stored at +2C to +8C.

Dosage and administration


Only trained healthcare workers should perform smallpox vaccination. One of
several techniques can be used to place a tiny volume of the reconstituted vaccine
on the skin of the lateral surface of the upper arm. Most commonly, a bifurcated
needle is dipped into a multidose container and then positioned vertically over
the skin, which is then punctured repeatedly with sufficient vigour to produce no
more than a trace of blood at the site.4-6,8
Personal protective equipment must be used while performing smallpox
vaccination.
Smallpox vaccines must not be injected subcutaneously, intramuscularly or
intravenously.
Intradermal inoculation with smallpox vaccine results in the formation of an
erythematous papule within 3 to 5days. It becomes a vesicle, then a pustule
reaching a maximum size of 1 to 2 cm in 8 to 12days, then scabs and separates
by 14 to 21days. When the procedure results in this circumscribed infection,
vaccination provides long-term protection against fatal disease. Furthermore,
vaccination very soon after exposure to smallpox markedly attenuates or
prevents clinical disease.4-6

Recommendations
The only current indication for vaccination in Australia is for workers using live
pox virus in recombinant gene research, in order to prevent infection at sites
of accidental inoculation. Currently, no vaccine is licensed for use in Australia;
however, information about sources of vaccines and their use should be obtained
from the Therapeutic Goods Administration, Canberra.4
Australian guidelines for smallpox outbreak, preparedness, response and
management may be found at http://www.health.gov.au/internet/wcms/
publishing.nsf/Content/health-pubhlth-publicat-others.htm.4

284 The Australian Immunisation Handbook 9th Edition

Contraindications
The vaccine is contraindicated in:4,5
people with diseases that cause impaired immunity such as human
immunodeficiency virus (HIV) infection, acquired immune deficiency
syndrome (AIDS), leukaemia, lymphoma, generalised malignancy,
agammaglobulinaemia,
those undergoing therapy with alkylating agents, antimetabolites, radiation
or large doses of steroids,
those who have ever been diagnosed with eczema, even if the condition is
mild or not presently active,
those with a history of neurological disorder,
women who are either pregnant or trying to become pregnant,
women who are breastfeeding,
children aged <1 year,
anyone living in a household with a member who has any of the conditions
listed above,
people with serious, life-threatening allergies to the antibiotics polymyxin
B, streptomycin, tetracycline or neomycin (this may depend on brand of
vaccine used),
those vaccinated in the past 30days with a live vaccine,
those with a history of cardiac disease, including:
previous myocardial infarction,
angina,
congestive heart failure,
cardiomyopathy,
valvular disease, including rheumatic heart disease,
stroke or transient ischaemic attack,
chest pain or shortness of breath with activity,
other heart conditions under the care of a doctor.

Individuals with acute or chronic skin conditions, such as atopic dermatitis,


impetigo and varicella-zoster (chickenpox and shingles), should not be
vaccinated until the condition resolves.
Individuals with eczema should live apart from recently vaccinated family
members who may have skin lesions.

Smallpox 285

3.20 Smallpox

Precautions4-6

Women should be advised to avoid pregnancy for 3months after smallpox


vaccination.
Anyone who receives a smallpox vaccination should not receive another live
vaccine for 1month afterwards.

Adverse events4-6,8
Smallpox vaccines have well described adverse events, which vary in frequency
according to the virus present in the seed stock. They include:
postvaccinal encephalitis (PVE) or encephalomyelitis (PVEM), a
demyelinating disease which occurs at a rate of 1 per 300000 vaccinations;
PVE is generally seen in those aged <1 year and PVEM in those aged
>2years;
progressive vaccinia (vaccinia gangrenosa) at the site of inoculation, in
vaccinees with immune impairment;
eczema vaccinatum, being vaccinial skin disease at sites of previous or
current eczema; occurs at a rate of about 1 in 26000 vaccinations;
generalised vaccinia, a self-limiting condition resulting from blood-borne
dissemination of the virus to other skin sites; more serious in people with
impaired immunity; occurs at a rate of 1 in 5000 vaccinations;
inadvertent inoculation of either the vaccinee or vaccinator in sites such as
the face, eyes or hands; occurs at a rate of 1 in 20000 primary vaccinations;
various skin rashes, usually self-limiting but can progress to Stevens-Johnson
Syndrome;
fetal vaccinia is rare (<50 reported cases), greatest risk occurs during the third
trimester;
cardiac adverse events including myocarditis, pericarditis and, possibly,
dilated cardiomyopathy.

Use in pregnancy
Smallpox vaccine is contraindicated in women who are either pregnant or trying
to become pregnant. Women should be advised to avoid pregnancy for 3months
after vaccination.

Vaccinia immune globulin4,5


Vaccinia immune globulin (VIG) is a sterile solution of the immunoglobulin
fraction of plasma containing antibodies to the vaccinia virus, from individuals
who were previously vaccinated with smallpox vaccine. VIG and the nucleoside
analogue active against poxviruses, cidofovir, may be used to treat vaccine
complications such as inadvertent inoculation of the eye or eyelid without
vaccinal keratitis, severe generalised vaccinia if patient is toxic, eczema

286 The Australian Immunisation Handbook 9th Edition

vaccinatum and progressive vaccinia. VIG is not indicated for treatment of


vaccinal keratitis or postvaccinal encephalitis.
VIG is contraindicated in those with a history of anaphylactic sensitivity to
thiomersal or to other humanised monoclonal antibodies.
There are currently limited stocks of VIG and cidofovir available in Australia.
Contact the Australian Government Department of Health and Ageing or
your State/Territory Health Department for further information regarding
these products (see Appendix 1, Contact details for Australian, State and Territory
Government health authorities and communicable disease control).

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

3.20 Smallpox

Smallpox 287

3.21 Tetanus
Bacteriology
Tetanus is caused by Clostridium tetani, a motile, non-capsulated, Gram-positive
rod that forms endospores. Spores of the bacillus are found in manured soil and
can enter wounds. Once in a wound site, the bacillus can grow anaerobically. C.
tetani produces a potent protein toxin which has 2 components, tetanospasmin (a
neurotoxin) and tetanolysin (a haemolysin).

Clinical features
Tetanus is an acute, often fatal, disease caused by the toxin produced by C. tetani.
The neurotoxin acts on the central nervous system to cause muscle rigidity
with painful spasms. The disease usually occurs after an incubation period of
3 to 21days (range 1 day to severalmonths), with a median time of onset after
injury of 10days. Generally, a shorter incubation period is associated with a
more heavily contaminated wound, more severe disease and a worse prognosis.
Generalised tetanus, the most common form of the disease, is characterised
by increased muscle tone and generalised spasms. Early symptoms and signs
include increased tone in the masseter muscles (trismus, or lockjaw), dysphagia,
stiffness or pain in the neck, shoulder and back muscles. Some patients develop
paroxysmal, violent, painful, generalised muscle spasms. A constant threat
during generalised spasms is reduced ventilation or apnoea or laryngospasm.
The patient may be febrile, although many have no fever; mental state is
unimpaired. Sudden cardiac arrest sometimes occurs, but its basis is unknown.
Other complications include pneumonia, fractures, muscle rupture, deep vein
thrombophlebitis, pulmonary emboli, decubitus ulcers and rhabdomyolysis.
Death results from respiratory failure, hypertension, hypotension or cardiac
arrhythmia.
Tetanus is rare in people who have received 5 doses of a tetanus-containing
vaccine (1 in 90 cases in the United Kingdom from 19842000).1 However,
individual cases have been reported2,3 and clinicians should consider
tetanus when there are appropriate symptoms and signs, irrespective of the
persons vaccination record. A high level of diagnostic awareness of tetanus
is particularly important in the elderly in industrialised countries, including
Australia, as most deaths occur in people over 70years of age, especially
women, and may be associated with apparently minor injury.1,4
Neonatal tetanus usually occurs as the generalised form and is usually fatal
if left untreated. It develops in children born to inadequately immunised
mothers, frequently after unsterile treatment of the umbilical cord stump. Its
onset generally occurs during the first 2weeks of life. Poor feeding, rigidity, and
spasms are typical features of neonatal tetanus.

288 The Australian Immunisation Handbook 9th Edition

Epidemiology

Effective protection against tetanus can be provided only by active immunisation.


Tetanus vaccine was introduced progressively into the childhood vaccination
schedule after World War II. The effectiveness of the vaccine was demonstrated
in that war; all Australian servicemen were vaccinated against tetanus and none
contracted the disease. As tetanus can follow apparently trivial, even unnoticed
wounds, active immunisation is the only certain protection.1 A completed course
of vaccination provides protection for manyyears.

Vaccines
The acronym DTPa, using capital letters, signifies child formulations of
diphtheria, tetanus and acellular pertussis-containing vaccines. The acronym
dTpa is used for adolescent/adult formulations which contain substantially
lesser amounts of diphtheria toxoid and pertussis antigens (see formulations).
Formulations for children aged <8years
Infanrix hexa GlaxoSmithKline (DTPa-hepB-IPV-Hib; diphtheriatetanus-acellular pertussis-hepatitis B-inactivated poliomyelitis vaccineHaemophilus influenzae type b (Hib)). The vaccine consists of both a 0.5mL
pre-filled syringe containing 30 IU diphtheria toxoid, 40 IU tetanus toxoid,
25g pertussis toxoid (PT), 25g filamentous haemagglutinin (FHA), 8g
pertactin (PRN), 10g recombinant HBsAg, 40 D-antigen units inactivated
polioviruses type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32
D-antigen units type 3 (Saukett) adsorbed onto aluminium hydroxide/
phosphate; phenoxyethanol as preservative; traces of formaldehyde,
polymyxin and neomycin and a vial containing a lyophilised pellet of 10g
purified Hib capsular polysaccharide (PRP) conjugated to 2040g tetanus
toxoid. The vaccine must be reconstituted by adding the entire contents of the
syringe to the vial and shaking until the pellet is completely dissolved. May
also contain yeast proteins.

Tetanus 289

3.21 Tetanus

In Australia, tetanus is rare, occurring primarily in older adults who have never
been vaccinated or were vaccinated in the remote past. There were 18 notified
cases of tetanus during 20012005, but 120 hospitalisations (July 2000June
2005) where tetanus was the principal diagnosis.4,5 This discrepancy suggests
under-notification. During 20012005, there were 2 deaths from tetanus.4,5 The
case-fatality rate in Australia is about 3%. Neonatal tetanus is a frequent cause of
infant mortality in parts of Asia, Africa and Latin America.

Infanrix-IPV GlaxoSmithKline (DTPa-IPV; diphtheria-tetanus-acellular


pertussis-inactivated poliomyelitis vaccine). Each 0.5mL pre-filled syringe
contains 30 IU diphtheria toxoid, 40 IU tetanus toxoid, 25g PT, 25g FHA,
8g PRN, 40 D-antigen units inactivated polioviruses type 1 (Mahoney),
8 D-antigen units type 2 (MEF-1) and 32 D-antigen units type 3 (Saukett)
adsorbed onto aluminium hydroxide; phenoxyethanol as preservative;
traces of formaldehyde, polymyxin and neomycin.
Infanrix Penta GlaxoSmithKline (DTPa-hepB-IPV; diphtheria-tetanusacellular pertussis-hepatitis B-inactivated poliomyelitis vaccine). Each
0.5mL pre-filled syringe contains 30 IU diphtheria toxoid, 40 IU tetanus
toxoid, 25g PT, 25g FHA, 8g PRN, 10g recombinant HBsAg, 40
D-antigen units inactivated polioviruses type 1 (Mahoney), 8 D-antigen
units type 2 (MEF-1) and 32 D-antigen units type 3 (Saukett) adsorbed onto
aluminium hydroxide/phosphate; phenoxyethanol as preservative; traces of
formaldehyde, polymyxin and neomycin. May also contain yeast proteins.
Formulations for people aged 8years
Adsorbed diphtheria-tetanus vaccine
ADT Booster Statens Serum Institut/CSL Biotherapies (dT; diphtheriatetanus, adult formulation). Each 0.5mL pre-filled syringe or monodose vial
contains 2 IU diphtheria toxoid and 20 IU tetanus toxoid adsorbed onto
0.5mg aluminium hydroxide.
Combination vaccines
Adacel Sanofi Pasteur Pty Ltd (dTpa; diphtheria-tetanus-acellular
pertussis). Each 0.5mL monodose vial contains 2 IU diphtheria toxoid, 20
IU tetanus toxoid, 2.5g PT, 5g FHA, 3g PRN, 5g pertussis fimbriae
(FIM) 2+3; 1.5mg aluminium phosphate; phenoxyethanol as preservative;
traces of formaldehyde.
Adacel Polio Sanofi Pasteur Pty Ltd (dTpa; diphtheria-tetanus-acellular
pertussis-inactivated poliomyelitis vaccine). Each 0.5mL monodose vial
contains 2 IU diphtheria toxoid, 20 IU tetanus toxoid, 2.5g PT, 5g
FHA, 3g PRN, 5g FIM 2+3, 40 D-antigen units inactivated polioviruses
type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32 D-antigen
units type 3 (Saukett); 1.5mg aluminium phosphate; phenoxyethanol
as preservative; traces of formaldehyde, polymyxin, neomycin and
streptomycin.

290 The Australian Immunisation Handbook 9th Edition

Boostrix-IPV GlaxoSmithKline (dTpa-IPV; diphtheria-tetanus-acellular


pertussis-inactivated poliomyelitis vaccine). Each 0.5mL pre-filled syringe
contains 2 IU diphtheria toxoid, 20 IU tetanus toxoid, 8g PT, 8g FHA,
2.5g PRN, 40 D-antigen units inactivated polioviruses type 1 (Mahoney),
8 D-antigen units type 2 (MEF-1) and 32 D-antigen units type 3 (Saukett)
adsorbed onto aluminium hydroxide/phosphate; traces of formaldehyde,
polymyxin and neomycin.
Tetanus vaccination stimulates the production of antitoxin, which protects
against the toxin produced by the organism. The immunogen is prepared by
treating a cell-free preparation of toxin with formaldehyde, thereby converting
it into the innocuous tetanus toxoid. Tetanus toxoid is usually adsorbed onto an
adjuvant, either aluminium phosphate or aluminium hydroxide, to increase its
immunogenicity. Antigens from Bordetella pertussis, in combination vaccines, also
act as an effective adjuvant.
Complete immunisation (5 doses) induces protective levels of antitoxin lasting
throughout childhood but, by middle age, about 50% of vaccinees have low
or undetectable levels.6-8 A single dose of tetanus toxoid produces a rapid
anamnestic response in such vaccinees.9-11
Tetanus toxoid is available in combination with other antigens. Production
of the previously available tetanus toxoid vaccine was discontinued by the
manufacturer in February 2006. Production of the previous DT (CDT vaccine),
registered for use in children <8years of age, ceased in June 2005. ADT Booster
can be used for the booster dose of dT in people aged 8years or, if necessary, for
the primary dT course (see Variations from product information below).

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.12 Store at
+2C to +8C. Protect from light. Do not freeze.

Dosage and administration


The dose of tetanus-containing vaccine is 0.5mL by IM injection.
Do not mix DTPa-containing vaccines, dTpa or dT vaccine with any other
vaccine in the same syringe, unless specifically registered for use in this way.

Tetanus 291

3.21 Tetanus

Boostrix GlaxoSmithKline (dTpa; diphtheria-tetanus-acellular pertussis).


Each 0.5mL monodose vial or pre-filled syringe contains 2 IU diphtheria
toxoid, 20 IU tetanus toxoid, 8g PT, 8g FHA, 2.5g PRN, adsorbed
onto 0.5mg aluminium hydroxide/phosphate; 2.5mg phenoxyethanol as
preservative. May contain traces of formaldehyde.

Recommendations
(i) Vaccination in childhood
Vaccination against tetanus is part of the National Immunisation Program
(NIP) schedule, with tetanus toxoid being given in combination with diphtheria
toxoid and acellular pertussis as DTPa vaccine. The recommended primary
course of vaccination is at 2, 4 and 6months of age. A booster dose of DTPa is
given at 4years of age. Immunity to tetanus will not be compromised before the
booster dose, as the serological response to the primary course of vaccination is
usually sufficient for thoseyears. A second booster, using the adolescent/adult
formulation, dTpa, at 1217years of age, is essential for maintaining immunity
to tetanus in adults. By the age of 17years, young adults should have received
5 doses of a tetanus toxoid-containing vaccine, and may have received an extra
dose if they have experienced a tetanus-prone wound during childhood.
For details on the management of children who have missed doses in the NIP
schedule, see Section 1.3.5, Catch-up.

(ii) Vaccination of adults


Booster vaccination
Routine 10-yearly booster doses in adults who have previously received 5 doses
of a tetanus-containing vaccine have not been recommended in Australia since
2000. All adults who reach the age of 50years and have not received a booster
dose of a tetanus-containing vaccine in the previous 10years should be given
dT or dTpa vaccine. This stimulates further production of circulating tetanus
antibodies at an age when waning of diphtheria and tetanus immunity is
commencing in the Australian population.6 The adolescent/adult formulation
dTpa is preferred, if not given previously, as it provides additional protection
against pertussis (see Chapter 3.14, Pertussis).
Primary vaccination
Where an adult has not received a primary course of tetanus toxoid previously,
3 doses of dT should be given, at minimum intervals of 4weeks, followed
by booster doses at 10 and 20years after the primary course. Give the first of
these doses as dTpa, to provide boosting to natural immunity from exposure
to pertussis, which is almost universal in unvaccinated adults. In the event that
dT vaccine is not available, dTpa can be used for all primary doses. However,
this is not recommended routinely because there are no data on the safety,
immunogenicity or efficacy of dTpa in multiple doses for primary vaccination.
Tetanus-prone wounds
Adults who have sustained injuries deemed to be tetanus prone should receive
a booster dose of dT, if more than 5years have elapsed since the last dose. In the
event that dT vaccine is not available, dTpa can be used (see Table 3.21.1 below).

292 The Australian Immunisation Handbook 9th Edition

(iii) Other people at special risk

Travellers to countries where health services are difficult to access should be


adequately protected against tetanus before departure. They should receive a
booster dose of dT, if more than 10years have elapsed since the last dose, or
dTpa if not given previously.

Tetanus-prone wounds
In the event of a tetanus-prone injury (defined below), a booster dose of vaccine
should be given if more than 5years have elapsed since the last dose. If there
is any doubt about the adequacy of previous tetanus immunisation, tetanus
immunoglobulin (see below) should be given as well as tetanus toxoid (see Table
3.21.1). In children <8years of age, this dose of vaccine should be given as DTPa
or a DTPa-combination vaccine, consistent with the childs vaccination history
and the NIP schedule. For details on the management of children who have
missed doses in the NIP schedule, see Section 1.3.5, Catch-up.
The definition of a tetanus-prone injury is not straightforward, as tetanus
may occur after apparently trivial injury, such as from a rose thorn, or with no
history of injury. However, there are certain types of wounds likely to favour the
growth of tetanus organisms. These include compound fractures, bite wounds,
deep penetrating wounds, wounds containing foreign bodies (especially wood
splinters), wounds complicated by pyogenic infections, wounds with extensive
tissue damage (eg. contusions or burns) and any superficial wound obviously
contaminated with soil, dust or horse manure (especially if topical disinfection
is delayed more than 4 hours). Reimplantation of an avulsed tooth is also a
tetanus-prone event, as minimal washing and cleaning of the tooth is conducted
to increase the likelihood of successful reimplantation.

Tetanus 293

3.21 Tetanus

Adults who were born in countries without adequate vaccination programs


may never have received primary vaccination against tetanus. Older adults may
have inadequate antitoxin levels, due to incomplete primary vaccination against
tetanus. Injecting drug users are at risk of tetanus, particularly if skin popping
is practised.13

General measures for treatment of tetanus-prone wounds14-19


Table 3.21.1: Guide to tetanus prophylaxis in wound management
History
of tetanus
vaccination

Time since
last dose

Type of wound

DTPa, DTPacombinations,
dT, dTpa, as
appropriate

Tetanus
immunoglobulin*
(TIG)

3 doses

<5years

All wounds

NO

NO

3 doses

510years

Clean minor
wounds

NO

NO

3 doses

510years

All other
wounds

YES

NO

3 doses

>10years

All wounds

YES

NO

<3 doses or
uncertain

Clean minor
wounds

YES

NO

<3 doses or
uncertain

All other
wounds

YES

YES

* The recommended dose for TIG is 250 IU, given by IM injection using a 21 gauge needle, as
soon as practicable after the injury. If more than 24 hours has elapsed, 500 IU should be given.
Individuals who have no documented history of a primary vaccination course (3 doses)
with a tetanus toxoid-containing vaccine should receive all missing doses. See Section 1.3.5,
Catch-up.

As an alternative to dT vaccine after a tetanus-prone wound, adults can receive


a single dose of dTpa vaccine to provide additional protection against pertussis
(providing they have not received a dose of dTpa previously).20
Whatever the immune status of an individual with a tetanus-prone wound,
local disinfection and, where appropriate, surgical treatment of tetanus-prone
wounds, must never be omitted. The use of antibiotics (such as penicillin or
metronidazole) for preventing infection is a matter for clinical judgement.
The recommended use of booster tetanus vaccines and the use of human tetanus
immunoglobulin are set out in Table 3.21.1. These should be administered as
soon as possible after the injury.

Tetanus immunoglobulin
Tetanus immunoglobulin (human) for intramuscular use
Tetanus Immunoglobulin-VF (TIG) CSL Bioplasma. 160mg/mL solution
of immunoglobulin from selected human plasma with high concentration of
antibodies to tetanus toxin, 250 IU.

294 The Australian Immunisation Handbook 9th Edition

(ii) The recommended dose for TIG is 250 IU by IM injection, to be given as soon
as practicable after the injury. If more than 24 hours have elapsed, 500 IU should
be given. A tetanus toxoid-containing vaccine should be given at the same time in
the opposite limb with a separate syringe, and arrangements should be made to
complete the full course of tetanus toxoid-containing vaccinations.
(iii) Because of its viscosity, TIG should be given to adults using a 21 gauge
needle. For children, it can be given slowly, using a 23 gauge needle.
(iv) For wounds not categorised as tetanus-prone, such as clean cuts that have
been treated appropriately, TIG is unnecessary.

Tetanus immunoglobulin (human) for intravenous use


Tetanus Immunoglobulin-VF (human, for intravenous use) CSL Bioplasma.
60mg/mL solution of immunoglobulin fraction of selected human plasma
with high concentration of antibodies to tetanus toxin, 4000 IU.
This product is used in the management of clinical tetanus. The recommended
dose is 4000 IU given by slow intravenous infusion. Detailed protocols for
administration of this product and management of adverse events should be
consulted if its use is contemplated.

Contraindications
The only absolute contraindications to tetanus vaccine are:
anaphylaxis following a previous dose of the vaccine, or
anaphylaxis following any vaccine component.
If an individual has a tetanus-prone wound and has previously had a severe
adverse event following tetanus vaccination, alternative measures, including the
use of human tetanus immunoglobulin, can be considered.

Precautions
In previously vaccinated people, administration of more than 1 dose of a tetanuscontaining vaccine in a 5-year period may provoke adverse events.

Adverse events
Mild discomfort or pain at the injection site persisting for up to a fewdays is
common. Uncommon general adverse events following dT vaccine include
headache, lethargy, malaise, myalgia and fever. Anaphylaxis, urticaria and
peripheral neuropathy very rarely occur (brachial neuritis occurs in 0.001% of

Tetanus 295

3.21 Tetanus

(i) TIG should be used for passive protection of individuals who have sustained
a tetanus-prone wound, where the person has not received 3 or more doses of
a tetanus toxoid-containing vaccine or where there is doubt about their tetanus
vaccination status. TIG provides immediate protection, for a period of 3 to 4weeks.

cases). For specific adverse events following combination vaccines containing


both tetanus and pertussis antigens, see Chapter 3.14, Pertussis.

Use in pregnancy
Refer to Chapter 2.3, Groups with special vaccination requirements, Table 2.3.1
Vaccinations in pregnancy.

Variations from product information


The product information for both Infanrix hexa and Infanrix Penta states that
these vaccines may be given as a booster dose at 18months of age. NHMRC
recommends that a booster dose of DTPa (or DTPa-containing vaccines) is not
necessary at 18months of age. However, DTPa-containing vaccine may be used
for catch-up of the primary schedule in children <8years of age.
The product information for Infanrix-IPV states that this vaccine may be used as
a booster dose for children 6years of age who have previously been vaccinated
against diphtheria, tetanus, pertussis and poliomyelitis. NHMRC recommends
that booster doses of DTPa and IPV be given at 4years of age; however, this
product may be used for catch-up of the primary schedule or as a booster in
children <8years of age.
The product information for ADT Booster states that this vaccine is indicated
for a booster dose only in children aged 5years and adults who have
previously received at least 3 doses of diphtheria and tetanus vaccines. NHMRC
recommends that, where a dT vaccine is required for any person 8years of
age, ADT Booster can be used, including for primary immunisation against
diphtheria and tetanus.
The product information for adolescent/adult formulations of dTpa-containing
vaccines states that these vaccines are indicated for booster doses only.
NHMRC recommends that, where dT is unavailable for the primary course,
dTpa can be used.
The product information for Adacel and Boostrix (adolescent/adult formulations
of dTpa) states that these vaccines are recommended for use in those aged
>10years. However, NHMRC recommends that they may be used in people
aged 8years. The product information also states that dTpa should not be
given within 5years of a tetanus toxoid-containing vaccine. However, NHMRC
recommends that dTpa vaccines can be administered at any time following
receipt of a diphtheria and tetanus toxoid-containing vaccine.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

296 The Australian Immunisation Handbook 9th Edition

3.22 Tuberculosis
Bacteriology
Tuberculosis (TB) is caused by organisms of the Mycobacterium tuberculosis
complex (M.TB complex), that are slow-growing, aerobic, acid-fast bacilli. The
M.TB complex consists of Mycobacterium tuberculosis, M. bovis, M. microti, M.
canetti and M. africanum.1 M. tuberculosis is the cause of almost all TB in Australia,
whereas M. bovis, M.canetti and M. africanum are rare.2

Clinical features

Most individuals infected with M. tuberculosis remain asymptomatic, but there is


a 10% lifetime risk of developing clinical illness, sometimes manyyears after the
original infection. Infants, the elderly and patients with impaired immunity due
to drugs or disease or as a result of adverse socioenvironmental circumstances
(eg. malnutrition, alcoholism) are more prone to rapidly progressive or
generalised infection.1,4

Epidemiology
The World Health Organization (WHO) declared tuberculosis a global emergency
in 1993, and recent reports have reaffirmed the threat to human health.5 About
1000 cases of TB are notified to Australian health authorities each year. The
annual notification rate for TB has been relatively stable at approximately 5 to
6 cases per 100000 population since 1985, and multi-drug resistance remains
rare, occurring in less than 2% of notified cases.2 Tuberculosis in animals (M.
bovis) has been eradicated by screening and culling programs. In Australia, most
TB cases (greater than 80%) occur in people born overseas, particularly in Asia,
southern and eastern European countries, the Pacific Islands, and north and subSaharan Africa. The rates of TB in the overseas-born population have been slowly
increasing over the past decade.3 High TB rates seen in people from Ethiopia,
Somalia and the Sudan reflect recent changes in the composition of Australias
migrant and refugee intake.3,6 Rates of TB are also high in Aboriginal and Torres
Strait Islander people and in Papua New Guineans living in some parts of
Australia.3,7

Tuberculosis 297

3.22 Tuberculosis

As infection is usually air-borne, lung disease is the most common form of


tuberculosis, accounting for approximately 60% of notified TB cases in Australia.3
Cough, fever, sweats, weight loss and haemoptysis are common symptoms
of pulmonary TB. TB lymphadenitis is the most common extrapulmonary
manifestation, but the disease can occur in any part of the body, including the
meninges, bone and kidneys. Disseminated disease (miliary TB) and meningeal
TB are the most serious forms, particularly in children.1

Patients with impaired immunity are at high risk of developing active TB if


they are infected with M. tuberculosis.4,5 Screening programs in Australia now
concentrate on those at high risk, including contacts of notified patients.

Vaccine
BCG vaccine Sanofi Pasteur Pty Ltd (freeze-dried live vaccine prepared
from an attenuated strain of Mycobacterium bovis). When reconstituted with
accompanying buffered saline diluent, vaccine contains between 832 x
106 colony forming units permL and monosodium glutamate 1.5% w/v.
Reconstituted vaccine provides about 10 adult or 20 infant doses.
BCG (Bacille Calmette-Gurin) vaccine is a suspension of live attenuated M. bovis.
Worldwide, there are many BCG vaccines available but they are all derived from
the strain propagated by the Institut Pasteur and first tested in humans in 1921.8
Protective efficacy ranges from 0 to 80% in controlled trials. The variation has
been attributed to differences in vaccine strains, local prevalence of (protective)
environmental mycobacteria, and host factors such as age at vaccination and
nutritional status. Geographic latitude and vaccine strains explain most of the
variation in efficacy. However, it should be noted that BCG is highly effective in
children, particularly those <5years of age, for whom it is primarily intended.
The efficacy of BCG in adolescents and adults is less.
BCG is primarily intended for children as meta-analyses have found the
protective efficacy for preventing serious forms of TB in this group is over
80%.9 Protective efficacy in all age groups is about 50%.10,11 An Australian study
reported a protective efficacy of 30%, at best, in school-aged children.12 Protective
efficacy is difficult to quantify and may vary from 10years to 50years.13,14 The
WHO does not recommend repeat vaccination.
In some studies, BCG has been shown to offer some protection against leprosy.15

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.16 Store
reconstituted vaccine at +2C to +8C or unreconstituted (freeze-dried) vaccine
in a freezer at 20C. Protect vaccine from light (sunlight or fluorescent). Store
diluent at +2C to +8C and do not freeze. Reconstituted BCG vaccine is very
unstable and should be discarded after one working session of 8 hours. Do not
freeze reconstituted BCG vaccine.

Dosage and administration


BCG vaccine is administered as a single dose by intradermal injection. It
should be given only by specially trained medical or nursing staff who are fully
conversant with the following procedures:

298 The Australian Immunisation Handbook 9th Edition

Tuberculin test all individuals, except infants <6months of age, before


vaccination. Read the test 48 to 72 hours later and, where 5 tuberculin units has
been given, give BCG only to those who have <5mm of induration.
Give 0.1mL of BCG to children and adults, and 0.05mL to infants
<12months of age.
Use a short (10mm) 2627 gauge needle with a short bevel. The risk of
spillage can be minimised by using an insulin syringe to which the needle is
already attached.
Wear protective eye-wear. The patient and parent holding the patient (if
patient is a small child requiring restraint) should also wear protective eyewear. Eye splashes may ulcerate, so if an eye splash occurs, wash the eye with
saline or water immediately.

Stretch the skin between a finger and thumb and insert the bevel into the
dermis, bevel uppermost, to a distance of about 2mm. The bevel should be
visible through the transparent epidermis.
If the injection is not intradermal, withdraw the needle and try again at a new
site. A truly intradermal injection should raise a blanched bleb of about 7mm
in diameter with the features of peau dorange. Considerable resistance will
be felt as the injection is given. If this resistance is not felt, the needle may be
in the subcutaneous tissues.
Advise the subject of adverse events which may follow the injection.
A tuberculin reaction induced by BCG usually ranges from 0 to 15mm, but
clinical trials have not shown a consistent relationship between the size of
tuberculin reactions and the protection provided. For this reason, tuberculin
skin testing of BCG vaccinees is not routinely recommended. Because of waning
hypersensitivity, most adults who were vaccinated with BCG in early childhood
will have a negative tuberculin test.
BCG is available from State/Territory tuberculosis services.

Response to BCG vaccination


A small red papule forms and eventually ulcerates, usually within 2 to 3weeks
of vaccination. The ulcer heals with minimal scarring over severalweeks. There
may be swelling and tenderness in local lymph nodes. Subjects who are given
BCG despite previous tuberculosis infection will experience an accelerated
response characterised by induration within 24 to 48 hours, pustule formation in
5 to 7days and healing within 10 to 15days.

Tuberculosis 299

3.22 Tuberculosis

Inject BCG into the skin over the region of the insertion of the deltoid muscle
into the humerus. This is just above the mid-point of the upper arm. This site
is recommended to minimise the risk of keloid formation. By convention, the
left upper arm is used wherever possible to assist those who subsequently
look for evidence of BCG vaccination.

Recommendations
(i) Given the low incidence of TB in Australia and the variable efficacy in adults,
BCG is not used in the general population.
(ii) BCG is recommended for the following:17
Aboriginal and Torres Strait Island neonates living in regions of high TB
incidence,
neonates born to parents with leprosy or a family history of leprosy,
children <5years of age who will be travelling to live in countries of high TB
prevalence for longer than 3months (WHO defines high-risk countries as
those with an annual incidence of TB in excess of 100 per 100000 population
see http://www.who.int/tb/en/),
embalmers,
healthcare workers involved in conducting autopsies.
(iii) State and Territory guidelines should be consulted for advice on vaccination
of the following groups of individuals:17
healthcare workers who may be at high risk of exposure to drug-resistant
cases,
neonates weighing <2.5kg,
children 5years and <16years of age who will be travelling or living for
extended periods in countries with a high prevalence of tuberculosis.

Contraindications
The use of BCG vaccine is contraindicated in the following:
individuals with impaired immunity due to HIV infection, corticosteroids
or other immunosuppressive agents, congenital immunodeficiencies and
malignancies involving bone marrow or lymphoid systems (because of the
risk of disseminated BCG infection) (see also Chapter 2.3, Groups with special
vaccination requirements),
individuals with a high risk of HIV infection where HIV antibody status is
unknown,
individuals with any serious illness including the malnourished,
individuals with generalised septic skin diseases and skin conditions such as
eczema, dermatitis and psoriasis,
pregnant women (BCG has never been shown to cause fetal damage, but use
of live vaccines in pregnancy is not recommended),
individuals who have previously had TB or a large (5mm) tuberculin
(Mantoux) reaction,
individuals with significant febrile illness (administer 1month from the time
of recovery).

300 The Australian Immunisation Handbook 9th Edition

Precautions
BCG should be deferred in the following:
neonates with a birth weight <2.5kg or those who may be relatively
malnourished,
neonates of mothers who are HIV-positive,
children who are currently on isoniazid preventive therapy for latent TB
infection (as the therapy can inactivate the BCG),
a 4-week interval should be allowed after administration of another live
vaccine (MMR, varicella [and MMRV when available], yellow fever vaccine)
unless given concurrently with the BCG.

About 5% (common) of vaccinees experience adverse events. 2.5% develop


injection site abscesses and 1% lymphadenitis. About 1% (uncommon) may
need medical attention including surgery as a result of the adverse event.18
Anaphylactoid reactions have also been reported. Gross local or generalised
infection can be treated with antituberculous drugs. Keloid formation can occur,
but the risk is minimised if the injection is not given higher than the level of the
insertion of the deltoid muscle into the humerus.

Use in pregnancy
Use of BCG in pregnancy is not recommended. BCG has never been shown to
cause fetal damage, but use of live vaccines in pregnancy is contraindicated.

The tuberculin skin test (TST)


(i) Hypersensitivity to tuberculin Purified Protein Derivative (PPD) follows
either natural infection with either M. tuberculosis or with other mycobacteria that
induce cross-reactivity, or BCG vaccination. The skin test is used (a) to detect past
infection for epidemiological purposes, (b) to detect latent TB infection (LTBI),
especially in contacts of TB patients, (c) as an aid in diagnosing disease due to TB,
and (d) as a pre-vaccination screen before BCG to prevent vaccine reactions.
(ii) Most tuberculin testing in Australia is performed using the Mantoux
technique. The PPD preparation for this test is currently supplied by Sanofi
Pasteur Pty Ltd. The product, Tubersol, comes in multidose vials and has 5
Tuberculin units (TU)/0.1mL (10 doses per 1mL vial). For routine testing, 0.1mL
of PPD (ie. a dose of 5 TU) is injected intradermally into the skin of the upper
third of the flexor surface of the left forearm, producing a peau dorange bleb
4 to 10mm in diameter. The reaction is examined 48 to 72 hours later, and the
diameter of the palpably indurated skin is measured across the long axis of the
forearm and recorded in millimetres. In certain circumstances, 2-step skin testing
may be required. It is used to detect individuals previously infected who may

Tuberculosis 301

3.22 Tuberculosis

Adverse events

test negative to tuberculin testing initially, but who show a strong reaction to
tuberculin if the same procedure is repeated 1 to 2weeks later. The 2-step test
is important to establish the baseline reaction when future tuberculin testing is
required as part of contact tracing or monitoring of high-risk groups. Detailed
information can be accessed in the Tubersol product information and relevant
State/Territory guidelines.
(iii) Erythema without induration should be disregarded. Strongly positive
reactions may be accompanied by skin necrosis, lymphangitis and regional
adenitis. Patients with a history of such strongly positive reactions to previous
testing should not be retested.
(iv) The reaction to PPD may be suppressed by recent surgery, sarcoidosis,
immunosuppressant drugs and illnesses, such as Hodgkins disease, lymphoma
and HIV infection that result in impaired immunity. The reaction also wanes
with increasing age, so that most adults vaccinated with BCG in childhood have
negative tuberculin reactions.
(v) The reaction to PPD may be unreliable for 4weeks after administration of
other live vaccines including MMR, varicella and yellow fever vaccines unless
given concurrently. The tuberculin skin test should be deferred for patients with
major viral infections or live-virus vaccination in the pastmonth. Oral typhoid
and oral polio (OPV is no longer used in Australia but may have been received
overseas) vaccines do not necessitate a delay in testing.
(vi) The use of the Heaf gun, a multiple puncture apparatus primed with highly
concentrated PPD, is not recommended.
(vii) Although BCG is not routinely recommended, it may be offered by some
States and Territories to their healthcare staff who should be made aware that
subsequent tuberculin skin testing may be difficult to interpret.
(viii) New interferon-gamma release assays (blood tests) using specific antigens
for M.tuberculosis are now available in some laboratories. Interferon-gamma
based assays are increasingly being used to diagnose latent TB and, particularly,
to distinguish latent TB from post-BCG vaccine skin reactivity. There are
relatively few data on the sensitivity and specificity of these tests in children,
particularly those <2years of age, or in people with impaired immunity.19

Variations from product information


The product information states that BCG should not be frozen. NHMRC advises
that BCG can be stored unreconstituted (freeze-dried) in a freezer at 20C.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

302 The Australian Immunisation Handbook 9th Edition

3.23 Typhoid
Bacteriology
Typhoid fever is a clinical syndrome caused by a systemic infection with
Salmonella enterica subspecies enterica serovar Typhi (S. Typhi). Paratyphoid fever,
caused by infection with S. enterica serovar Paratyphi A or B, is similar to, and
often indistinguishable from, typhoid fever. The two infections are collectively
known as enteric fever; there is no vaccine against paratyphoid fever.
NB. S. Paratyphi B biovar Java does not cause typhoid-like enteric fever but
rather causes a typical gastroenteritis; it can be acquired through handling
aquarium fish.1

Clinical features
Typhoid fever has a usual incubation period of 7 to 14days (range 3 to 60days).2
Although clinical presentations of typhoid fever can be quite variable, a typical
case presents with a low-grade fever, dull frontal headache, malaise, myalgia,
anorexia and a dry cough.2 The fever tends to increase as the disease progresses;
constipation (more typically diarrhoea in young children), abdominal tenderness,
relative bradycardia and splenomegaly are common. Complications occur in 10
to 15% of patients and tend to occur in patients who have been ill for >2weeks.
The more important complications include gastrointestinal bleeding, intestinal
perforation and typhoid encephalopathy.2

Because travellers are likely to seek medical advice relatively early in the illness,
severe typhoid fever with complications is rarely seen in this group of patients.
Nevertheless, travellers can still become chronic carriers of S. Typhi.

Epidemiology
Humans are the sole reservoir of S. Typhi. It is shed in the faeces of those acutely
ill and those who are chronic asymptomatic carriers of the organism; transmission
usually occurs via the ingestion of faecally-contaminated food or water.
The vast majority of typhoid fever cases occur in less-developed countries where
poor sanitation, poor food hygiene and untreated drinking water all contribute
to endemic disease with moderate to high incidence and considerable mortality.3
Geographic regions with high incidence (>100 cases per 100000 population

Typhoid 303

3.23 Typhoid

Relapse occurs in 5 to 10% of patients, usually 2 to 3weeks after the initial fever
resolves. Chronic asymptomatic biliary carriage of S. Typhi occurs in up to 5% of
patients with typhoid fever, even after treatment. Chronic carriage is defined by
the continued shedding of the organism for >1year, and is a public health risk
(eg. if a carrier works in the food industry).2

per year) include the Indian subcontinent, most southeast Asian countries and
several south Pacific nations, including Papua New Guinea.
In developed countries, typhoid fever is predominantly a travel-related disease,
with a considerably greater risk following travel to the Indian subcontinent
than to other regions.4,5 Those who travel to endemic regions to visit friends and
relatives (ie. immigrants who travel to their former homelands) appear to be at
considerably greater risk of acquiring typhoid fever than other travellers.4,5 There
are approximately 50 to 80 cases of typhoid fever reported in Australia each year,
with most following travel to regions with endemic disease.

Vaccines
Vivotif Oral CSL Biotherapies/Berna Biotech Ltd (oral live attenuated
typhoid vaccine). Each enteric-coated capsule contains not less than 2 x 109
viable organisms of attenuated S. Typhi strain Ty21a Berna. 3 capsules in a
blister pack.
Typherix GlaxoSmithKline (purified Vi capsular polysaccharide vaccine).
Each 0.5mL pre-filled syringe contains 25g Vi polysaccharide of S. Typhi;
phenol as preservative; phosphate buffer. 10 dose packs are also available.
Typhim Vi Sanofi Pasteur Pty Ltd (purified Vi capsular polysaccharide
vaccine). Each 0.5mL pre-filled syringe contains 25g Vi polysaccharide of
S. Typhi; phenol as preservative; phosphate buffer.
Combination vaccine that includes both typhoid and hepatitis A (see
Chapter 3.5, Hepatitis A)
Vivaxim Sanofi Pasteur Pty Ltd (inactivated hepatitis A virus and typhoid
Vi capsular polysaccharide). Supplied in a unique dual-chamber syringe
which enables the 2 vaccines to be mixed just before administration. Each
1.0mL dose of mixed vaccine contains 160 ELISA units of inactivated
hepatitis A virus antigens, 25g purified typhoid capsular polysaccharide;
0.3mg aluminium hydroxide; 2.5 L phenoxyethanol; formaldehyde; traces
of neomycin and bovine serum albumin.
The attenuated non-pathogenic S. Typhi strain Ty21a was derived by chemical
treatment attenuation of a wild-type strain. Attenuated features of Ty21a include
the absence of the enzyme, UDP-galactose-4-epimerase, and the Vi capsular
polysaccharide antigen (an important virulence determinant of S. Typhi). These
features partially contribute to the non-pathogenicity and, therefore, the safety of
the oral live vaccine.6
The oral vaccine Ty21a strain cannot be detected in faeces more than 3days after
administration of the vaccine. It stimulates vigorous secretory intestinal IgA and
cell-mediated immune responses.6 Clinical trials, with different formulations of

304 The Australian Immunisation Handbook 9th Edition

the vaccine and with a variety of schedules, have been undertaken in several
countries (Egypt, Chile, Indonesia) with endemic typhoid fever. These have
documented varying degrees of protection against the disease.2,6
The parenteral Vi polysaccharide vaccine is produced by fermentation of the
Ty2 strain followed by inactivation with formaldehyde, and then extraction of
the polysaccharide from the supernatant using a detergent.6 The vaccine elicits
prompt serum IgG anti-Vi responses in 85 to 95% of adults and children >2years
of age. The vaccine has also been used in clinical trials in endemic regions (Nepal,
South Africa, China), indicating moderate protection against typhoid fever.2,6
Neither the oral nor the parenteral vaccines have been studied in prospective
clinical trials in travellers to endemic regions. Because many travellers do not
have any naturally-acquired immunity, the protection conferred through typhoid
vaccination may be less than that documented in the clinical trials mentioned
above. However, there is circumstantial evidence that the vaccines do provide
protection to travellers to endemic regions,4,5 and that 3-yearly revaccination is
necessary to prolong the protection.7

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.8 Store all
typhoid vaccines at +2C to +8C. Protect from light. Do not freeze.
Because the vaccinee will be responsible for looking after the course of the oral live
attenuated vaccine, details of how it should be transported (from the pharmacy to
the home) and stored in the refrigerator (at home) must be carefully explained.

Oral live attenuated vaccine


The vaccine is registered for use in individuals 6years of age; it is presented
in a pack of 3 capsules. The dose (a whole capsule) is the same for both adults
and children.
It may be administered at the same time as either OPV (no longer used in
Australia), or yellow fever vaccine.6 It may also be given concurrently with
mefloquine or with atovaquone/proguanil combination (Malarone).
The vaccination schedule consists of 1 capsule of vaccine ondays 1, 3, and 5,
taken 1 hour before food. The capsule must be swallowed whole with water and
must not be chewed since the organisms can be killed by gastric acid. Do not give
the vaccine concurrently with antibiotics, or other drugs that are active against
Salmonellae. If possible, antibiotics and other relevant drugs should be delayed for
3days after the last dose of the vaccine.
A fourth capsule taken on day 7 has been shown (in a single study9) to result in a
lower incidence of typhoid fever than 3 doses. However, the use of a fourth dose
requires partial use of a second pack and, therefore, may involve considerable
extra expense.

Typhoid 305

3.23 Typhoid

Dosage and administration

Oral live attenuated typhoid vaccine should be separated from the


administration of inactivated oral cholera vaccine by an interval of at least 8
hours (see Precautions below).

Parenteral Vi polysaccharide vaccines


Both vaccines (Typherix and Typhim Vi) are registered for use in individuals
2years of age; the dose (0.5mL) is the same for both adults and children. (The
dose of the combined Vi polysaccharide and hepatitis A vaccine is 1mL.) The
vaccines are given by IM injection.

Booster doses
The optimal timing of revaccination against typhoid fever is uncertain and,
therefore, international recommendations can vary considerably.2,5,6
However, if continued exposure to S. Typhi exists (such as occurs with
either prolonged travel or residence in an endemic region) it is reasonable to
recommend a dose of the parenteral vaccine 3years after the initial primary
parenteral vaccination. If a 3-dose schedule of the oral live attenuated vaccine
was used initially, a repeat 3-dose course can be given after 3years; if a 4-dose
schedule of the oral vaccine was used initially, a repeat 4-dose course can be
given after 5years.6

Recommendations
Typhoid vaccination is recommended for all travellers 2years of age going to
endemic regions, where food hygiene may be suboptimal and drinking water
may not be adequately treated. Travellers include the military. Vaccination
should be completed at least 2weeks before travel.
Individuals travelling to endemic regions to visit friends and relatives are
probably at considerable risk of acquiring typhoid fever, and vaccination is
strongly recommended for them.
NB. Travellers must also be advised about personal hygiene, food safety and
about drinking boiled or bottled water only. They should be advised that raw (or
undercooked) shellfish, salads, cold meats, untreated water and ice (in drinks)
are all potentially high-risk, as are short (day) trips away from higher quality
accommodation venues.
Laboratory personnel routinely working with S. Typhi should also be considered
for vaccination.

306 The Australian Immunisation Handbook 9th Edition

Contraindications
The only absolute contraindications to typhoid vaccines are:
anaphylaxis following a previous dose of a typhoid vaccine, or
anaphylaxis following any component of a particular typhoid vaccine.

Oral live attenuated vaccine


The oral live attenuated vaccine should not be administered
to pregnant women; parenteral Vi polysaccharide vaccine should be used
instead;
to individuals with impaired immunity, including those known to be infected
with HIV; parenteral Vi polysaccharide vaccine should be used instead; or
to individuals taking antibiotics; parenteral Vi polysaccharide vaccine should
be used instead.

Parenteral Vi polysaccharide vaccines


There are no other contraindications to these vaccines.

Precautions
There should be an interval of at least 8 hours between the administration of the
inactivated oral cholera and oral typhoid vaccines, as the buffer in the cholera
vaccine may affect the transit of the capsules of oral typhoid vaccine through the
gastrointestinal tract.

The oral live attenuated vaccine should not be administered to children <6years
of age; parenteral Vi polysaccharide vaccine should be used instead in children
25years of age.

Adverse events
Typhoid vaccines, both oral and parenteral, are associated with very few adverse
events and, when adverse events do occur, they tend to be mild and transient.10

Oral live attenuated vaccine


Abdominal discomfort, diarrhoea, nausea, vomiting and rashes have occasionally
been reported.

Parenteral Vi polysaccharide vaccines


Local adverse events such as erythema, swelling and pain at the injection site
are very common (10 to 20%). Systemic adverse events are common and include
fever (3%), malaise and nausea.

Typhoid 307

3.23 Typhoid

The Vi polysaccharide vaccines should not be administered to children <2years


of age.

Public health management of typhoid fever


Typhoid fever is a notifiable disease in all States and Territories in Australia.
Upon notification, the relevant public health authority should ascertain the likely
source of infection, and determine if the case has an occupation (eg. as a food
handler or carer of children, the elderly or of those with impaired immunity)
where there might be the potential for the spread of S. Typhi.11 Cases in such
occupations should be excluded from work until 2 consecutive faecal samples,
collected aweek apart after the completion of antibiotic treatment, are negative
for S.Typhi.11 Cases not in such occupations should also be proved to have
cleared the organism from 2 consecutive faecal samples, but they can return to
work once they have recovered and any diarrhoea has ceased.
All those who have been in close contact with a case of typhoid fever in the 30days
before the clearance of S. Typhi from the cases faeces should have their occupation
assessed (as above). Contacts in risk occupations should be excluded from work
until 2 faecal samples, collected at least 24 hours apart, have proved negative for
S.Typhi.11 Contacts not in such occupations can remain at work, but should have at
least 1 faecal sample collected. Further instructions about the management of cases
of typhoid fever, and their contacts, should be obtained from State/Territory public
health authorities (see Appendix 1, Contact details for Australian, State and Territory
Government health authorities and communicable disease control).

Use in pregnancy
The oral live attenuated vaccine should not be given in pregnancy. However,
pregnancy is not a contraindication to vaccination with a parenteral Vi
polysaccharide vaccine. Refer to Chapter 2.3, Groups with special vaccination
requirements, Table 2.3.1 Vaccinations in pregnancy.

Variations from product information


The product information for the oral live attenuated vaccine does not mention
the use of a 4-dose course of the vaccine for either primary or booster
vaccination.
Although NHMRC considers pregnancy a contraindication to the oral live
attenuated typhoid vaccine, the product information does not include pregnancy
among the listed contraindications.
The Typhim Vi product information recommends a booster dose every 2 to
3years. However, revaccination with a dose of Vi polysaccharide vaccine 3years
after an initial dose is recommended by NHMRC.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

308 The Australian Immunisation Handbook 9th Edition

3.24 Varicella
Virology
Varicella-zoster virus (VZV) is a DNA virus within the herpes virus family.1
Primary infection with VZV causes varicella (chickenpox). Following primary
infection, VZV establishes latency in the dorsal root ganglia. Reactivation of the
latent virus manifests as herpes zoster (shingles)2 (see Chapter 3.26, Zoster).

Clinical features
Varicella is a highly contagious infection spread by air-borne transmission of
droplets from the upper respiratory tract or from the vesicle fluid of the skin
lesions of varicella or zoster infection.1 Varicella is usually a mild disease of
childhood. However, complications occur in approximately 1% of cases.3 It is
more severe in adults and in individuals of any age with impaired immunity, in
whom complications, disseminated disease, and fatal illness can occur.1
The average incubation period is 14 to 16days (range 1021days), but may
be longer in those with impaired immunity, especially after receipt of zoster
immunoglobulin (ZIG).2 The period of infectivity is from 48 hours before the
onset of rash until crusting of all lesions has occurred. A short prodromal
period of 1 to 2days may precede the onset of the rash, especially in adults.1,2
In otherwise healthy children, skin lesions usually number between 200 and
500.1,2 Acute varicella may be complicated by secondary bacterial skin infection,
pneumonia, acute cerebellar ataxia (1 in 4000 cases), aseptic meningitis,
transverse myelitis, encephalitis (1 in 100000 cases), and thrombocytopenia. In
rare cases, it involves the viscera and joints.1

Reactivation of latent VZV as a result of waning cellular immunity results in


herpes zoster (HZ), a localised vesicular rash. HZ can occur at any age, but
is more common in older adults and individuals with impaired immunity.
Complications may include post-herpetic neuralgia, and disseminated zoster
with visceral, central nervous system and pulmonary involvement1 (see Chapter
3.26, Zoster).

Varicella 309

3.24Varicella

Congenital varicella syndrome has been reported after varicella infection in


pregnancy and may result in skin scarring, limb defects, ocular anomalies,
and neurologic malformations.1,4 There is a higher risk to the fetus if maternal
infection occurs in the second trimester compared with infection in the
first trimester (1.4% vs 0.55%).5 Infants with intrauterine exposure also risk
developing herpes zoster in infancy (0.81.7%) with the greatest risk following
exposure in the third trimester.4 Severe neonatal varicella infection can result
from perinatal maternal varicella.6 The onset of varicella in pregnant women
from 5days before delivery to 2days after delivery is estimated to result in
severe varicella in 17 to 30% of their newborn infants.7

There is no specific therapy for uncomplicated varicella infection. Antiviral


therapy is used in the treatment of complicated or severe disease or varicella in
people with impaired immunity. An increased risk of Reye syndrome following
varicella infection has been reported in association with aspirin or other salicylate
use8-12 (see Precautions below). Aspirin or other salicylates should not be used in
the management of varicella infection.

Epidemiology
In an unimmunised population in temperate climates, the annual number
of cases of varicella approximates the birth cohort.13 Tropical regions have a
higher proportion of cases in adults. Approximately 5% of cases are subclinical.
A serosurvey conducted in 19971999 found that 83% of the Australian
population were seropositive by 1014years of age.14 Before the introduction
of a varicella vaccination program in Australia there were about 240000 cases,
1500 hospitalisations and an average of 7 to 8 deaths each year from varicella in
Australia.15-17 The highest rates of hospitalisation occur in children <4years of age.18
In the USA, universal varicella vaccination since 1995 has resulted in a decline in
varicella disease by 85% and hospitalisations have declined by 70 to 88%.19-21 The
greatest decline in hospitalisation rates has been in 04-year-olds, who were most
likely to be vaccinated. However, a reduction in hospitalisation rates also occurred
in older children and adults, due to herd immunity.19 Surveillance of varicella
and HZ in the USA, conducted between 1992 and 2002, demonstrated that, as
vaccination coverage increased to 65% in 2002, the incidence of varicella decreased
by 65% across all age groups, and the incidence of HZ remained stable.22

Vaccines
Live attenuated varicella vaccine (VV) is currently available as a monovalent
vaccine. It is anticipated that quadrivalent combination vaccines containing
measles, mumps, rubella and varicella vaccines (MMRV) will be available in the
near future (see Chapter 3.11, Measles for information on MMRV vaccines). All
available varicella-containing vaccines are derived from the Oka VZV strain, but
have some genetic differences.23
Monovalent VVs have been available in Australia since 2000, and from
November 2005, a single dose of VV has been funded under the NIP for
all children at 18months of age, with a catch-up dose funded for children
10<14years of age.24 At the time of implementation of a universal varicella
vaccination program in Australia, a single dose was considered adequate for
protection of infants and children <14years of age. However, recent data from
the USA suggests that a second dose of varicella-containing vaccine in children
is optimal to provide an immune response more like natural infection, reducing
the risk of vaccine failure and increasing population immunity.7 Vaccine failure is
known as breakthrough varicella and is defined as a case of wild-type varicella
42days post vaccination. A majority of cases of breakthrough varicella are

310 The Australian Immunisation Handbook 9th Edition

mild with fewer lesions than natural infection. However, breakthrough varicella
infections can be contagious.25
Post-licensure studies in the USA have estimated the effectiveness of 1 dose of
VV in children to be 80 to 85% against any disease and 95 to 98% against severe
varicella.25-29 Although earlier data suggested persistence of immunity in most
healthy vaccinees,1 recent long-term data from the United States has shown
that, >5years after vaccination, rates of vaccine failure increased by 2.6 times
in children who received only 1 dose of vaccine, compared with those who had
received the vaccine within 5years.30 Data from a randomised controlled trial
in varicella-negative children 12months to 12years of age, comparing 1 with 2
doses of VV over a 10-year period, showed significantly higher protection with
2 doses (94.4% vs 98.3%).31 Based on current evidence, 2 doses of a varicellacontaining vaccine in children from 12months of age will minimise the risk of
breakthrough varicella (see Recommendations below).
Healthy adolescents (14years of age ) and adults require 2 doses of varicella
vaccine, 1 to 2months apart, as the response to a single dose of VV decreases
progressively as age increases and is insufficient to provide adequate protection.32
Monovalent varicella vaccines
Varilrix GlaxoSmithKline (lyophilised preparation of live attenuated Oka
strain of varicella-zoster virus). Each 0.5mL monodose of the reconstituted,
lyophilised vaccine contains not less than 103.3 plaque-forming units of
attenuated varicella-zoster virus; human serum albumin; lactose; neomycin;
polyalcohols. Single and 10 pack of monodose vials also available.
Varivax Refrigerated CSL Biotherapies/Merck & Co Inc (lyophilised
preparation of live attenuated Oka/Merck strain of varicella-zoster virus).
Each 0.5mL monodose of the reconstituted, lyophilised vaccine contains
not less than 1350 plaque-forming units; 18mg sucrose; 8.9mg gelatin;
3.6mg urea; 0.36mg monosodium glutamate; residual components of
MRC-5 cells; trace amounts of neomycin and fetal bovine serum from
MRC-5 culture media. Single and 10 pack of monodose vials also available.

Transport, storage and handling

Varilrix store at +2C to +8C. Protect from light. Do not freeze. After
reconstitution, use within 90 minutes at ambient temperature or for up to 8 hours
at +2C to +8C.

Varicella 311

3.24Varicella

Varicella vaccines are less stable than other commonly used live viral vaccines,
and the storage temperature requirements are critical. Available monovalent VVs
have different storage requirements.

Varivax Refrigerated store at +2C to +8C. Protect from light. Do not freeze.
After reconstitution, use within 30 minutes at ambient temperature (+20C to
+25C) to maintain potency.
Transport according to National Vaccine Storage Guidelines: Strive for 5.33

Dosage and administration


The dose of monovalent VV is 0.5mL, administered by SC injection.
If VV is given at the same time as MMR, it should be given using separate
syringes and injection sites. MMR and monovalent VV should not be mixed
before injection.
VV can be given at the same time as other vaccines (including MMR, DTPa,
hepatitis B and MenCCV), using separate syringes and injection sites. If VV is not
given simultaneously with other live viral parenteral vaccines (eg. MMR), they
should be given at least 4weeks apart (see Precautions below).

Recommendations
(i) Children
It is recommended that at least 1 dose of a varicella-containing vaccine be given
to all non-immune children from the 2nd year of life to <14years of age. Children
in this age group with a reliable history of varicella infection, either by confident
clinical diagnosis or with laboratory confirmation, may be considered immune
and do not require vaccination.
Routine varicella-containing vaccine should be administered as follows (and as
per Table 3.24.1):
One dose of monovalent VV at 18months of age.
When MMRV vaccines become available, 1 dose of varicella-containing
vaccine should be given as MMRV at 12months of age.
The change in the recommended age of administration of varicella vaccine
is influenced by moving the second dose of MMR to 18months of age,
and the anticipated availability of MMRV vaccines in the near future. The
available evidence now suggests that the administration of varicella vaccine
at the earlier age of 12months, compared with 18months, does not reduce
vaccine effectiveness or lead to increased rates of breakthrough varicella.34
Administration of varicella vaccine at 12months of age will provide earlier
protection from varicella. However, until MMRV vaccines are available in
Australia, it is recommended that administration of monovalent VV at 18months
of age continue to avoid schedule crowding (4 injections) at 12months of age.
Receipt of 2 doses of varicella-containing vaccine provides increased protection
and minimises the chance of breakthrough varicella in children.31 However, at
this time, routine administration of 2 doses of varicella-containing vaccine at
<14years of age is not included on the NIP schedule. If parents/carers wish to

312 The Australian Immunisation Handbook 9th Edition

minimise the risk of breakthrough varicella, a second dose of varicella-containing


vaccine to children <14years of age is recommended (see Vaccines above).
When available, use of MMRV at 18months of age is a suitable means to provide
a second dose of varicella-containing vaccine. (For further information, see
also Chapter 3.11, Measles.) The minimum approved interval between doses of
varicella-containing vaccine in children <14years of age is 4weeks.
Table 3.24.1: Recommendations for varicella vaccination with monovalent VV
(currently available), and once MMRV vaccines are available
Monovalent varicella
vaccine

12months

18months

MMR

MMR + VV

MMRV when available MMRV

Catch-up requirements*

MMR

No requirement for
varicella catch-up
Use MMRV at 18months for
children who have not yet received
at least 1 dose of varicella vaccine

* If catch-up is required for MMR, see Chapter 3.11, Measles.


Give in separate syringes and at separate injection sites (preferably the other arm).
When available, use of MMRV at 18months of age is a suitable means to provide a second
dose of varicella-containing vaccine.

(ii) Adolescents (14years of age) and adults


Vaccination is recommended in non-immune adolescents (14years of age) and
adults. Immune responses are reduced in adolescents and adults compared
with young children.32,35 Therefore, adolescents and adults must receive 2 doses
of VV to achieve adequate protection from varicella. The 2 doses should be
administered at least 4weeks apart. However, a longer interval between vaccine
doses is acceptable. Lack of immunity to varicella should be based on a negative
history of previous varicella infection and can be supplemented by serological
testing (see Serological testing before varicella vaccination below).
VV is particularly indicated for those in the following categories:
non-immune people in high-risk occupations where exposure to varicella
is likely (such as healthcare workers, teachers and workers in childcare
centres) (see Section 2.3, Table 2.3.6 Recommended vaccinations for those at risk of
occupationally acquired vaccine-preventable diseases),36

seronegative women immediately after delivery,


non-immune parents of young children, and
non-immune household contacts of all ages of people with impaired
immunity.

Varicella 313

3.24Varicella

non-immune women before pregnancy to avoid congenital or neonatal


varicella,

MMRV vaccines are not recommended for use in adolescents and adults because
data are currently available only for children 12years of age.

(iii) Serological testing before varicella vaccination


Vaccination is well tolerated in previously infected individuals and can be
administered if there is uncertainty regarding immunity. Serological testing
before varicella vaccination of children with a reliable history of varicella
infection, either by confident clinical diagnosis or with laboratory confirmation,
is not warranted. Reliable history of varicella infection correlates highly with
serological evidence of immunity.37,38 Those who have an uncertain history
should be considered susceptible and offered vaccination.
In adolescents and adults with a negative history of varicella infection,
serological testing before vaccination is more likely to be cost-effective,
as a majority of those with a negative history may be immune.36,39
However, vaccination can proceed without testing (provided there are no
contraindications), as the vaccine is well tolerated in seropositive people.

(iv) Serological testing after varicella vaccination


Testing to check for seroconversion after varicella vaccination is not
recommended. Commercially available laboratory tests are not always
sufficiently sensitive to detect low antibody levels following vaccination and, in
addition, the presence of detectable antibody shortly after vaccination does not
necessarily indicate complete immunity to varicella.40,41

(v) Post-exposure prophylaxis and outbreak control


Several studies have shown that VV is effective in preventing varicella infection,
particularly moderate to severe disease, following exposure. This is generally
successful when given within 3days, and up to 5days, after exposure, with
earlier administration being preferable.42-46 Vaccination of exposed individuals
during outbreaks has also been shown to prevent further cases and control
outbreaks (see also The public health management of varicella below). When
available, vaccination with MMRV in children 12months to 12years of age could
be used for vaccination in this setting if MMR vaccination is also indicated.
In the event of an outbreak, seek advice from local public health authorities
before proceeding with vaccination of a large number of individuals (see
Appendix 1, Contact details for Australian, State and Territory Government health
authorities and communicable disease control).

(vi) Household contacts of people with impaired immunity


Vaccination of household contacts of people with impaired immunity is strongly
recommended. This recommendation is based upon evidence that transmission
of varicella vaccine virus strain is extremely rare and it is likely to cause only
mild disease that can be treated with acyclovir. This compares with the relatively
high risk of severe disease in people with impaired immunity following

314 The Australian Immunisation Handbook 9th Edition

exposure to wild-type varicella-zoster virus.41,47 If vaccinees develop a rash, they


should cover the rash and avoid contact with people with impaired immunity
for the duration of the rash. Zoster immunoglobulin (ZIG) need not be given
to an immune impaired contact of a vaccinee with a rash because the disease
associated with this type of transmission (should it occur) would be expected to
be mild.

(vii) Vaccination of healthcare workers (HCW)


(Refer to Table 2.3.6 Recommended vaccinations for those at risk of occupationally
acquired vaccine-preventable diseases)
Pre-exposure vaccination of HCWs:
A HCW with a negative or uncertain history of varicella infection should
undergo serological testing. If seronegative, vaccination should be offered in
a 2-dose schedule48 (see Recommendations (ii) above).
If a rash develops during the 6weeks after administration of the vaccine,
the HCW should cover the rash and be reassigned to duties that require no
patient contact, or placed on sick leave.48 Reassignment or leave should be
only for the duration of the rash48 (see Variations from product information
below).
VV-associated rash may be atypical and may not be vesicular (see Adverse
events below). A VV-associated rash is likely to occur in less than 5% of
vaccinees, and to last for less than 1week.49,50
Testing to check for seroconversion after VV is not recommended (see
Serological testing after varicella vaccination above).
Post-exposure management of HCWs:
If a previously vaccinated HCW is exposed to varicella, assume immunity
and report exposure. A vaccinated HCW should watch for a rash for 3weeks
after exposure and report to the nominated infection control officer should
a rash develop. If HCW vaccinees develop a rash, cover the rash, reassign
duties (no patient contact) or place on sick leave until no new lesions appear
and all lesions have crusted.

Varicella 315

3.24Varicella

If a HCW is exposed to varicella and is unvaccinated and has a negative


or uncertain history of varicella infection, offer vaccination. This is usually
effective in preventing the development of varicella if given within 3days,
and up to 5days, after exposure. In situations where facilities for rapid
testing are available, it may be possible to identify those with pre-existing
immunity before vaccination. However, serological testing should not delay
vaccination beyond the recommended 3 to 5days after exposure. Vaccination
in the absence of serological results is acceptable (see Serological testing after
varicella vaccination above).

If the HCW is vaccinated after exposure, as above, he/she can work but
should watch daily for any rash for 6weeks after exposure. Note that the
VV-associated rash may be atypical, maculopapular and non-vesicular.
If a varicella-exposed and vaccinated HCW develops a rash following
vaccination, this may be due to either wild-virus or vaccine-strain varicellazoster virus (see Adverse events below). In the event of a rash after
vaccination, cover the rash, reassign duties (no patient contact) or place on
sick leave until no new lesions appear and all lesions have crusted.
If an exposed non-immune HCW does not accept vaccination, reassign duties
or place on sick leave fromdays 10 to 21 from the time of first exposure.

Contraindications
(i) Allergy to vaccine components
Varicella vaccination is contraindicated where there has been:
anaphylaxis following a previous dose of any of the varicella vaccines, or
anaphylaxis following any vaccine component.

(ii) Pregnant women


VV should not be given during pregnancy and vaccinees should not become
pregnant for 28days after vaccination. Since wild-type VZV poses only a very
small risk to the fetus, the risk to the fetus of the attenuated VV virus, if any,
should be even lower. Data from a registry, established in the USA to monitor the
maternal-fetal outcomes of pregnant women who were inadvertently administered
VV 3months before or at any time during pregnancy, revealed that no birth defects
compatible with congenital varicella syndrome occurred in 254 known pregnancy
outcomes.51,52 The rate of occurrence of congenital anomalies from prospective
reports in the registry was similar to what is reported in the general USA
population (3.2%) and the anomalies showed no specific pattern or target organ.
A non-immune pregnant household contact is not a contraindication to
vaccination with VV of a healthy child or adult in the same household. The
benefit of reducing the exposure to varicella by vaccinating healthy contacts of
non-immune pregnant women outweighs any theoretical risks of transmission of
vaccine virus to these women.
Data on the use of MMRV vaccines in individuals >12years of age are not available.

(iii) People with impaired immunity


Varicella-containing vaccines are contraindicated in subjects with primary or
acquired impaired immunity, including:
people with impaired immunity due to HIV/AIDS. Vaccination with live
attenuated vaccine can result in a more extensive vaccine-associated rash
or disseminated infection in individuals with AIDS.53 However, varicella
vaccination of asymptomatic or mildly symptomatic HIV-infected children

316 The Australian Immunisation Handbook 9th Edition

may be considered (see Table 2.3.4 Immunological categories based on age-specific


CD4 counts and percentage of total lymphocytes). Since studies have not been
performed using combination MMRV vaccines in asymptomatic or mildly
symptomatic HIV-infected children, it is recommended that only MMR and
monovalent VV be considered for use in such children;54
people with conditions in which normal immunological mechanisms may be
impaired;
people suffering from malignant conditions of the reticuloendothelial system
(such as lymphoma, leukaemia, Hodgkins disease); and
people receiving high-dose systemic immunosuppressive treatment, such as
general radiation, x-ray therapy or oral corticosteroids. Varicella-containing
vaccines are contraindicated in those taking high-dose oral corticosteroids
(in children equivalent to either >2mg/kg per day prednisolone (20mg per
day total) for >1week or >1mg/kg per day for >4weeks) (see Section 2.3.3,
Vaccination of individuals with impaired immunity due to disease or treatment).
NHMRC also recommends that children who have been receiving high-dose
systemic steroids for 2weeks or more may be vaccinated after steroid therapy
has ceased for at least 1month.55 (See also Chapter 2.3, Groups with special
vaccination requirements and Chapter 3.11, Measles).

Precautions
(i) Vaccination with MMR
If VV is not given simultaneously with other live viral parenteral vaccines (eg.
MMR), they should be given at least 4weeks apart.

(ii) Vaccination after immunoglobulin or blood products


NHMRC recommends that varicella-containing vaccines should not be given
for between 3 and 9months after the administration of immunoglobulincontaining blood products. The interval between receipt of the blood product and
vaccination depends on the amount of antibody in each product, and is indicated
in Table 2.3.5 Recommended intervals between either immunoglobulins or blood
products and MMR, MMRV or varicella vaccination. For further information, see
Section 2.3.5, Vaccination of patients following receipt of other blood products including
blood transfusions, and Variations from product information below.

After vaccination with varicella-containing vaccines, immunoglobulin-containing


products should not be administered for 3weeks unless the benefits exceed those
of vaccination. If immunoglobulin-containing products are administered within
this interval, the vaccinee should either be revaccinated later at the appropriate
time following the product, as indicated in Table 2.3.5, or tested for immunity
6months later and then revaccinated if seronegative.

Varicella 317

3.24Varicella

(iii) Administration of immunoglobulin or bloodderived products after vaccination

(iv) Long-term aspirin or salicylate therapy


Individuals receiving long-term salicylate therapy should be vaccinated if
indicated, as the benefit is likely to outweigh any possible risk of Reye syndrome
occurring after vaccination. Natural varicella infection and salicylate use has
been associated with an increased risk of developing Reye syndrome. However,
there have been no reports of an association between Reye syndrome and
varicella vaccination (see Variations from product information below).

Adverse events
Adverse events following administration of VV are generally mild and well
tolerated.56 Fever >39C has been observed in 15% (common) of healthy
children, but this was comparable to that seen in children receiving placebo.55
In adults and adolescents, fever has been reported in 10% (common) of VV
vaccinees. Injection site adverse events (pain, redness or swelling) are the
most common adverse events reported after varicella vaccination, occurring
in 7 to 30% (common to very common) of vaccinees, but are generally well
tolerated.2,56 Slightly higher rates of fever were observed in the clinical trials
of MMRV vaccines, as compared with giving MMR and monovalent varicella
vaccine at the same time but at separate sites.57,58 It is recommended that
parents/vaccine recipients be advised about possible symptoms, and given
advice for reducing fever, including the use of paracetamol for fever in the
period 5 to 12days after vaccination.
A maculopapular or papulovesicular rash may develop after vaccination
(usually within 5 to 26days). Rashes typically consist of 2 to 5 lesions and
may be generalised (35%, common), or occur at the injection site (35%,
common).55 Most varicelliform rashes that occur within the first 2weeks
after vaccination are due to wild-type VZV, with median onset 8days
after vaccination (range 124days), while vaccine-strain VZV rashes occur
at a median of 21days after vaccination (range 542days).59 (See also
Transmission of vaccine virus below.)
No serious adverse events were reported from pre-licensure trials of
VV.1 A post-licensure study reported that serious adverse events, such as
encephalitis, ataxia, thrombocytopenia and anaphylaxis, were reported
following vaccination at a rate of 2.9 per 100000 doses distributed. However,
this does not necessarily imply a causal relationship.41
Transmission of vaccine virus to contacts of vaccinated individuals is rare. In
the USA, where more than 56 million doses of VV were distributed between
1995 and 2005, there have been only 6 well documented cases of transmission
of the vaccine-type virus from 5 healthy vaccinees.55,60 Contact cases have
been mild and associated with a rash in the vaccinee.55,60-62

318 The Australian Immunisation Handbook 9th Edition

Risk of herpes zoster (shingles)


Herpes zoster (HZ) has been reported rarely in vaccine recipients and has been
attributed to both the vaccine strain and to wild-type varicella virus reactivation.59
The risk of developing HZ is currently thought to be lower after vaccination than
after natural varicella virus infection, and reported cases have been mild.2 HZ is
uncommon before the age of 45years, and incidence increases with age.63 Rates
of herpes zoster in children 09years of age after natural VZV infection were
estimated to be 30 to 74 per 100000 per year.64,65 Vaccination results in a lower rate
of zoster with a rate of 22 per 100000 person-years reported in a 9-year follow-up
of 7000 varicella vaccinated children (Jane Seward, US Centers for Disease Control
and Prevention (CDC), personal communication) (see Chapter 3.26, Zoster).

Zoster immunoglobulin
High-titre zoster immunoglobulin (ZIG) is available from the Australian Red
Cross Blood Service on a restricted basis for the prevention of varicella in highrisk subjects. ZIG has no proven use in the treatment of established varicella or
zoster infection. ZIG must be given early in the incubation period (within 96
hours of exposure). ZIG is highly efficacious, but is often in short supply. Normal
human immunoglobulin (NHIG) can be used for the prevention of varicella if
ZIG is unavailable.
Zoster immunoglobulin should be given only by IM injection.
Zoster Immunoglobulin-VF (human) CSL Bioplasma (160mg/mL
immunoglobulin (IgG) preparation from human plasma containing high
levels of antibody to the varicella-zoster virus). Vials contain 200 IU, with
the actual volume stated on the label on the vial. Also contains glycine.

The public health management of varicella


Significant exposure is defined as living in the same household as a person with
active varicella or HZ, or direct face-to-face contact with a person with varicella
or HZ for at least 5 minutes, or being in the same room for at least 1 hour. In the
case of varicella infection, the period of infectivity is from 48 hours before the
onset of rash until crusting of all lesions has occurred.

ZIG should be given to individuals in the following categories within 96 hours of


significant exposure to either varicella or HZ:
Pregnant women who are presumed to be susceptible to varicella infection. If
practicable, they should be tested for varicella-zoster antibodies before ZIG is
given.4

Varicella 319

3.24Varicella

Immunocompetent varicella contacts should be tested for varicella-zoster


antibodies. However, this should not delay ZIG administration after initial
contact with a case.

ZIG must be given to neonates whose mothers develop varicella from 7 or


fewerdays before delivery to 2days after delivery, as the neonatal mortality
without ZIG is up to 30% in this setting. ZIG must be given as early as
possible in the incubation period.
ZIG should be given to neonates exposed to varicella in the firstmonth of life, if
the mother has no personal history of infection with VZV and is seronegative.
Premature infants (born at <28weeks gestation or <1000 g birth weight)
exposed to VZV while still hospitalised should be given ZIG regardless of
maternal history of varicella.
Patients suffering from primary or acquired diseases associated with cellular
immune deficiency, and those receiving immunosuppressive therapy. While
it is recommended that varicella contacts with impaired immunity be tested
for varicella-zoster antibodies, this should not delay ZIG administration,
preferably within 96 hours and up to 10days after initial contact with a case.66,67
NB. If a contact with impaired immunity is shown to have recent evidence of detectable
antibodies, it is not necessary to give ZIG, as its administration will not significantly
increase varicella-zoster antibody titres in those who are already positive. Note that
varicella-zoster antibodies detected in patients who have been transfused or who have
received intravenous immunoglobulin in the previous 3months may be passively
acquired and transient.
The following dose schedule is recommended for ZIG administration:
Table 3.24.2: Zoster immunoglobulin-VF (ZIG) dose based on weight
Weight of patient (kg)

Dose (IU)

010

200

1130

400

>30

600

A dose of ZIG may be repeated if a second exposure occurs more than 3weeks
after the first dose of ZIG. However, testing for varicella antibodies is also
recommended (see above). Normal human immunoglobulin can be used for the
prevention of varicella if ZIG is unavailable (see Chapter 3.8, Immunoglobulin
preparations). Patients receivingmonthly high-dose intravenous NHIG are likely
to be protected and probably do not require ZIG if the last dose of NHIG was
given 3weeks or less before exposure.

Use in pregnancy
Varicella vaccine is contraindicated during pregnancy (see Contraindications
(ii) Pregnant women above, and Chapter 2.3, Groups with special vaccination
requirements, Table 2.3.1 Vaccinations in pregnancy).
Pregnancy should be avoided for at least 28days after vaccination.

320 The Australian Immunisation Handbook 9th Edition

Variations from product information


Varilrix vaccine is approved for use in healthy children from 9months of age.
NHMRC recommends that routine vaccination of children against varicella
should occur at 12months of age.
Varilrix and Varivax Refrigerated are registered for use as 2 doses of 0.5mL
(12months apart) in adolescents 13years of age and adults. NHMRC
recommends a single dose of varicella vaccine for children <14years of age.
In adults and adolescents where 2 doses of vaccine are required, the product
information for Varilrix states that the second dose should be given at least
6weeks after the first. NHMRC recommends that the second dose may be given
at least 4weeks after the first dose.
For both varicella vaccines, the product information states that pregnancy should
be avoided for 3months after vaccination. NHMRC recommends that pregnancy
be avoided for at least 28days after vaccination.
For both varicella vaccines, the product information recommends that vaccinees
should avoid contact with people with impaired immunity for up to 6weeks
after vaccination. NHMRC recommends that healthcare worker vaccinees should
be reassigned to duties that involve no direct patient contact or be placed on sick
leave only if a rash develops, and that the period of leave or reassignment should
be only for the duration of the rash (not for the 4 to 6weeks stated in the product
information) (see also Vaccination of healthcare workers (HCW) above).
For both varicella vaccines, the product information states that salicylates should
be avoided for 6weeks after varicella vaccination, as Reye syndrome has been
reported following the use of salicylates during natural varicella infection.
NHMRC recommends that non-immune individuals receiving long-term
salicylate therapy should receive VV as the benefit is likely to outweigh any
possible risk of Reye syndrome occurring after vaccination.
The product information for Varivax Refrigerated recommends delaying
vaccination for 5months after receipt of NHIG by IM injection or blood
transfusion. The NHMRC recommends that VV should not be given for at least
3months in subjects who have received immunoglobulin-containing blood
products according to the intervals contained in Table 2.3.5.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

Varicella 321

3.24Varicella

The dosage of ZIG recommended in the product information differs with that in
Table 3.24.2, which has been revised in order to minimise wastage of ZIG.

3.25Yellow fever
Virology
Yellow fever is a viral haemorrhagic fever caused by a flavivirus that is
transmitted by mosquitoes. Aedes aegypti, a highly domesticated mosquito
found throughout the tropics, is the vector responsible for person-to-person
transmission of the yellow fever virus in urban and inhabited rural areas in
endemic countries.

Clinical features
The clinical spectrum of yellow fever varies from a non-specific febrile illness
to a fatal haemorrhagic fever.1 After an incubation period of 3 to 6days, the
disease begins abruptly with fever, prostration, myalgia and headache. The
patient appears acutely ill with congestion of the conjunctivae; there is an intense
viraemia during this period of infection which lasts 3 to 4days.1 This may be
followed by the period of remission in which the fever and symptoms settle
over 24 to 48 hours during which the virus is cleared by immune responses.1
Approximately 15 to 25% of patients may then relapse with a high fever,
vomiting, epigastric pain, jaundice, renal failure and haemorrhage: the period
of intoxication.1 These complications can be severe, and reflect the viscerotropic
nature of the yellow fever virus (its ability to infect the liver, heart and
kidneys). The case-fatality rate varies widely, but can be as high as 20% in local
populations.

Epidemiology
Yellow fever occurs in tropical regions of Africa and Central and South America.
In both regions the virus is enzootic in rainforest monkeys and canopy mosquito
species; sporadic human cases occur when people venture into these forests
(sylvatic or jungle yellow fever).1
In moist savannah regions in Africa, especially those adjacent to rainforests, tree
hole-breeding Aedes mosquito species are able to transfer yellow fever virus from
monkeys to people and then between people, leading to small-scale outbreaks
(intermediate yellow fever).
Aedes aegypti occurs in both heavily urbanised areas and settled rural areas in
tropical Africa and the Americas.1 Epidemics of urban yellow fever occur when
a viraemic individual (with yellow fever) infects local populations of Ae. aegypti;
such epidemics can be large and very difficult to control. Although Ae. aegypti
also occurs throughout much of tropical Asia and Oceania (including north
Queensland), yellow fever has never been reported from these regions.

322 The Australian Immunisation Handbook 9th Edition

The risk of susceptible travellers acquiring yellow fever varies considerably


with season, location, duration of travel and utilisation of mosquito avoidance
measures. There have been at least 6 reported cases of yellow fever, all fatal, in
unvaccinated travellers to Africa and South America since 1996.3

Vaccine
Stamaril Sanofi Pasteur Pty Ltd (live attenuated yellow fever virus
(17D-204 strain) lyophilised vaccine). Each 0.5mL monodose of
reconstituted, lyophilised vaccine contains not less than 1000 mouse LD50
units; lactose; sorbitol. May contain traces of egg proteins. The vaccine is
supplied as a single dose ampoule with 0.5mL diluent syringe.
Yellow fever vaccine is a live, freeze-dried preparation of the 17D attenuated
strain of yellow fever virus cultured in, and harvested from, embryonated
chicken eggs. The vaccine contains neither antibiotics nor preservatives, and does
not contain gelatin.
Vaccination elicits protective levels of neutralising antibodies in approximately
90% of adult vaccinees by day 14, and in virtually all by day 28.4 Immunity
is long-lasting and perhaps life-long; regardless, revaccination is required
after 10years under International Health Regulations for a valid International
Certificate of Vaccination. Because the vaccine produces a transient very low
level viraemia in healthy adult recipients, they cannot serve as a source of
infection for mosquitoes.4
Although the efficacy of the yellow fever vaccine has never been determined in
prospective clinical trials, there is considerable observational evidence that it is
very effective in preventing the disease.4

Transport, storage and handling


Transport according to National Vaccine Storage Guidelines: Strive for 5.5 The
vaccine and diluent should be stored at +2C to +8C. Do not freeze. Protect from
light. The vaccine should be used within 1 hour of reconstitution.

Yellow fever 323

3.25Yellow fever

Although yellow fever is undoubtedly markedly under-reported, it is clear that


there has been a considerable increase in the reported number of outbreaks,
and therefore cases, of yellow fever in recent decades.2 Most of this increase has
been in Africa, particularly in West African countries.2,3 Between 2000 and 2004,
18 of the 25 countries at risk in Africa reported cases of yellow fever, with 13 of
the 14 countries at risk in West Africa reporting cases.3 During this time, West
Africa experienced 5 epidemics of urban yellow fever, 3 of which affected capital
cities (Abidjan (Cte dIvoire, 2001), Conakry (Guinea, 2002), Dakar and Touba
(Senegal, 2002) and Bobo-Dioulasso (Burkina Faso, 2004)).3

Dosage and administration


The vaccine can be given to those 9months of age. The dose is 0.5mL of
reconstituted vaccine regardless of age, given by either IM or SC injection.

Recommendations
Yellow fever is considered to be endemic in 32 African and 11 Central and South
American countries (Table 3.25.1). Of these, 25 African and 9 South American
countries are currently considered at risk because they have reported cases since
1950; of particular concern are the countries in West Africa (Table 3.25.1).3,6
Table 3.25.1: Yellow fever endemic countries
West African countries

Other endemic countries

Benin *

Angola*

Guyana*

Burkina Faso *

Bolivia*

Kenya*

Cte dIvoire *

Brazil*

Niger

Gambia *

Burundi

Panama

Ghana *

Cameroon*

Peru*

Guinea *

Central African Republic*

Rwanda

Guinea-Bissau *

Chad

Sao Tome and Principe

Liberia *

Colombia*

Somalia

Mali *

Congo*

Sudan*

Mauritania *

Democratic Republic
of the Congo*

Suriname*

Nigeria *

Ecuador*

Trinidad and Tobago

Senegal *

Equatorial Guinea*

Uganda*

Sierra Leone *

Ethiopia*

United Republic of Tanzania

Togo *

French Guiana*

Venezuela*

Gabon*
* Countries with cases reported since 1950.
Travellers who will visit only the cities of Cuzco and Machu Picchu do not need vaccination.

The yellow fever vaccine is recommended for:


those 9months of age travelling or living in any country in West Africa (see
Table 3.25.1), regardless of where they will be in that country,
those 9months of age travelling or living outside urban areas of all other
yellow fever endemic countries (see Table 3.25.1), and
laboratory personnel who routinely work with yellow fever virus.

324 The Australian Immunisation Handbook 9th Edition

As well as the above recommendations, many countries outside the endemic


regions require evidence of vaccination for those travellers arriving from endemic
countries. Because Ae.aegypti exists in many of these non-endemic countries,
there is a potential for yellow fever virus transmission; a listing of these countries
is available in an annex at www.who.int/ith/en/.
Travellers >1 year of age arriving into Australia within 6days of leaving a yellow
fever endemic country are required to have a valid International Certificate of
Vaccination against Yellow Fever (see below). Such travellers who do not have
a valid certificate are placed under quarantine surveillance until 6days have
passed after leaving the endemic country. Quarantine surveillance does not
place restrictions on the travellers movements, but does require prompt medical
assessment should the traveller develop relevant symptoms.
Yellow fever vaccine can be administered only by Yellow Fever Vaccination
Centres approved by the relevant State or Territory health authorities. Each
yellow fever vaccination is to be recorded in an International Certificate of
Vaccination against Yellow Fever; the certificate must include the vaccinees
name and signature (or the signature of a parent or guardian if the vaccinee is a
child), and the signature of a person approved by the relevant health authority.
The date of the vaccination must be recorded in day-month-year sequence with
themonth written in letters, and the official stamp provided by the State or
Territory health authority must be used. The certificate becomes valid 10days
after vaccination, and remains valid for 10years.
NB. People with a true contraindication to yellow fever vaccine (see below) who
intend to travel to endemic countries (as recommended above) should obtain a
letter from a doctor, clearly stating the reason for withholding the vaccine. The
letter should be formal, signed and dated, and on the practices letterhead.

Contraindications
(i) Known anaphylactic sensitivity
The yellow fever vaccine is contraindicated in those who have had either:
anaphylaxis following a previous dose, or
anaphylaxis following any of the vaccine components.
In particular, the vaccine is contraindicated if there is a known anaphylaxis to eggs.

Yellow fever 325

3.25Yellow fever

All those travelling or living in endemic countries should be informed that the
mosquito vectors of yellow fever usually bite during the day, and they should be
advised of the necessary mosquito avoidance measures. These include the use of
insect repellents, coils and sprays, the use of mosquito nets (preferably those that
have been treated with an insecticide), and adequate screening of residential (and
work) premises.

(ii) Infants
Yellow fever vaccine is contraindicated in infants <9months of age.

(iii) Altered immune status


As with all live virus vaccines, the yellow fever vaccine should not be given to
people with impaired immunity due to either disease or medical treatment (see
Chapter 2.3, Groups with special vaccination requirements).

(iv) Thymus disorders


People with a history of any thymus disorder, including myasthenia gravis,
thymoma, thymectomy and DiGeorge syndrome, should not be given the yellow
fever vaccine.

Precautions
(i) Adults 60years of age
The risk of severe adverse events following yellow fever vaccine is
considerably greater in those aged 60years than in younger adults.7,8
Adults 60years of age should be given yellow fever vaccine only if they intend
to travel to endemic countries (as recommended above) and they have been
informed about the (albeit very low) risks of developing a severe complication.

(ii) Pregnancy
As with all live virus vaccines, unless there is a risk of exposure to the virus,
yellow fever vaccine should not be given to pregnant women. Pregnant women
should be advised against going to the rural areas of yellow fever endemic
areas (and to urban areas of West African countries as well). However, pregnant
women who insist on travelling, against medical advice, to endemic countries
should be vaccinated.
The yellow fever vaccine has been given to considerable numbers of pregnant
women4,9 with no evidence of any adverse outcomes. Therefore, women
vaccinated in early pregnancy can be reassured that there is no evidence of risk
to themselves and very low (if any) risk to the fetus; there are no grounds for a
termination of pregnancy.4

(iii) Administration of other vaccines on the same day


The administration of other live virus vaccines (MMR, varicella vaccine [and
MMRV when available]) should be on the same day as yellow fever vaccine, or
separated by a 4-week interval. Other vaccines relevant to travel can be given
with, or at any time before or after, yellow fever vaccine.

326 The Australian Immunisation Handbook 9th Edition

Adverse events
(i) Common adverse events

(ii) Immediate hypersensitivity reactions


Immediate hypersensitivity reactions, including anaphylaxis, following yellow
fever vaccine are very rare with an incidence of less than 1 in 1 million and occur
principally in people with anaphylactic sensitivity to eggs.4 Although it has
been suggested that an anaphylactic sensitivity to gelatin (added as a stabiliser
to some yellow fever vaccines) may also precipitate anaphylaxis following
vaccination,11 Stamaril does not contain gelatin.

(iii) Vaccine-associated neurotropic adverse events


At least 25 cases of meningoencephalitis following yellow fever vaccination
have been reported.4 However, 15 of these cases occurred in the 1950s in infants
7months of age; following recommendations in the early 1960s not to vaccinate
young infants, the incidence of vaccine-associated meningoencephalitis declined
considerably.4 Nevertheless, these adverse events, albeit very rare, still occur in
adults; the risk is greater in vaccinees 60years of age.7,8

(iv) Vaccine-associated viscerotropic adverse events


Recently, an apparently very rare (and often fatal) complication, characterised
by multiorgan system failure, has been recognised following yellow fever
vaccination; it appears that overwhelming infection with the 17D vaccine-virus is
responsible for these viscerotropic adverse events.4 Up to October 2004, 25 such
cases had been reported worldwide; the exact incidence is unknown but may be
less than 1 in 400000 doses of vaccine.4
Vaccine-associated viscerotropic adverse events do not appear to be caused by
altered virulence of the vaccine-virus, but rather appear to be related to host
factors. Although cases have occurred in younger people, it is apparent that
the risk is increased with advanced age, in particular in those aged 60years.7,8
Another host factor associated with an increased risk of a viscerotropic adverse
event is pre-existing thymus disease. Four of the 25 reported cases had a history
of thymic tumour and thymectomy, both uncommon conditions.12

Use in pregnancy
Pregnant women, who insist on travelling to either a West African country, or
outside urban areas of any other endemic country, should be vaccinated (see
Precautions above).

Yellow fever 327

3.25Yellow fever

Adverse events following yellow fever vaccine are generally mild. Vaccinees
often report mild headaches, myalgia and low-grade fevers or other minor
symptoms for 5 to 10days post vaccination. In clinical trials in which symptoms
are actively elicited, up to 25% of vaccinees report mild adverse events and up to
1% curtail regular activities.4,10

Variations from product information


The product information states that pregnancy is a contraindication to the yellow
fever vaccine. However, NHMRC recommends that pregnant women who insist
on travelling, against medical advice, to endemic countries, should be vaccinated.
The product information suggests that a 0.1mL test dose of yellow fever vaccine
can be used intradermally to assess an individual with suspected allergy to the
vaccine. However, NHMRC states that (with the exception of Q fever vaccine)
test doses should never be used.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

328 The Australian Immunisation Handbook 9th Edition

3.26Zoster (herpes zoster)


Virology
Varicella-zoster virus (VZV) is a DNA virus that is a member of the herpesvirus
family. Herpes zoster (HZ) or shingles is caused by reactivation of latent VZV
which typically resides in the dorsal root or trigeminal ganglia following primary
infection with VZV.1 Primary infection with VZV is known as varicella or
chickenpox.1

Clinical features

Post-herpetic neuralgia (PHN), the most frequent debilitating complication


of HZ, is a neuropathic pain syndrome that persists or develops after the
dermatomal rash has healed or persists longer than 3months after the onset of
the rash.5,6 Other complications associated with HZ, such as ophthalmic disease
and neurological complications, may occur depending on the site of reactivation.
Rarely, disseminated HZ may develop with visceral, central nervous system, and
pulmonary involvement. Disseminated disease is more common in people with
impaired immunity.4 Dermatomal pain without the appearance of rash is also
documented (zoster sin herpte). Antiviral therapy, intiated within 3days after
the onset of HZ, reduces the severity and duration of HZ and may reduce the
risk of developing PHN.7-11 However, in many cases, PHN may persist foryears,
and may be refractory to treatment.12

Epidemiology
HZ occurs most commonly with increasing age (>50years), impaired immunity,
and a history of varicella in the first year of life. The lifetime risk of reactivation
of VZV causing HZ is estimated to be approximately 20 to 30%, and it affects
half of those who live to 85years.1,13-15 Second attacks of HZ occur in less than
5% of immunocompetent individuals, but are more frequent in individuals with
impaired immunity.2,16 Internationally, average incidence rates of HZ in the total
population vary from 130 to 405 per 100000 person-years depending on the
study population.17,18 Australian data on the incidence of HZ in the community
are limited. However, using general practice and other data, approximately

Zoster (herpes zoster) 329

3.26Zoster (herpes zoster)

Reactivation of VZV causing HZ is thought to be due to a decline in cellular


immunity to the virus, and presents clinically as a unilateral vesicular rash in
a dermatomal distribution in the majority of cases. It is often painful and lasts
10 to 15days.1,2 A prodromal phase occurs 48 to 72 hours before the appearance
of the lesions in 80% of cases.3 Associated symptoms may include headache,
photophobia, malaise, and an itching, tingling, or severe pain in the affected
dermatome.2,4 In the majority of patients, HZ is an acute and self-limiting disease.
However, complications can occur, especially with increasing age.

5 cases per 1000 population (range 3.38.3 per 1000) are estimated to occur
annually.19-21
Overall, an estimated 13 to 26% of HZ patients develop complications.
Complications occur more frequently with increasing age, and with impaired
immunity.22,23 PHN occurs infrequently in young people but, in patients >50years
of age, it complicates HZ in 25 to 50% of cases.1
Modelling the outcomes of the introduction of a universal infant vaccination
schedule for varicella has predicted a rise in the incidence of HZ based on the
assumption that exposure to VZV boosts immunity.24 It has been suggested that
an increase in HZ incidence rates (in the population previously infected with
wild-type VZV) will occur for approximately 40years, based on a varicella
vaccine coverage rate of 90% and boosting preventing HZ for an average of
20years.21,24 Surveillance in the USA has not suggested a change in the incidence
of HZ since the introduction of a universal varicella vaccination program in
1995.18 The incidence of HZ in children vaccinated with varicella vaccine is less
than in those infected with wild-type virus.1,25
In most states of Australia, surveillance for varicella and HZ is currently being
implemented in order to track the burden of disease from VZV before and after
the introduction of the varicella vaccination program.26 South Australia has
conducted passive surveillance of varicella and HZ since January 2002.

Vaccine
A frozen formulation of a live attenuated zoster vaccine is currently registered
in Australia but is not marketed. The zoster vaccine is formulated from the same
VZV strain (Oka/Merck) as the licensed varicella (chickenpox) vaccine, Varivax,
but is of higher potency (at least 14 times greater). The higher viral titre in the
zoster vaccine is required in order to elicit a sufficient cellular immune response
in adults who usually remain seropositive to VZV but have declining cellular
immunity with increasing age. The licensed varicella vaccines are not suitable for
use in the prevention of zoster in older people.
Several randomised placebo-controlled trials conducted during the clinical
development of the zoster vaccine confirmed that a vaccine at potencies above
19400 plaque forming units (PFU) stimulated both VZV-specific antibodies and a
cell-mediated immune response.27,28 A large randomised, double-blind, placebocontrolled efficacy and safety trial of the zoster vaccine (known as the Shingles
Prevention Study) was conducted among 38546 primarily healthy adults aged
60years, and demonstrated that the zoster vaccine was safe and generally well
tolerated. Vaccination significantly reduced the burden of both HZ and PHN.27
Overall, compared with placebo, vaccination reduced the incidence of HZ by
51.3% (95% CI: 44.257.6%), the incidence of PHN by 66.5% (95% CI: 47.579.2%),
and the burden of illness associated with pain from HZ by 61.1% (95% CI:
51.169.1%) over a median of 3.12years of follow-up.27

330 The Australian Immunisation Handbook 9th Edition

No product is currently marketed in Australia (see Appendix 3, Products registered


in Australia but not currently available).

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

3.26Zoster (herpes zoster)

Zoster (herpes zoster) 331

Appendix 1: Contact details for


Australian, State and Territory
Government health authorities and
communicable disease control
Australian Government health authorities
Australian Government
Health

02 6289 1555

Australian Childhood
Immunisation Register
enquiries (ACIR)

1800 653 809


(This phone number can also be used by
vaccination providers to obtain information on the
vaccination history of individual children).
ACIR email: acir@medicareaustralia.gov.au
ACIR Internet site: www.medicareaustralia.gov.au;
click on Health Care Providers, then Programs & Services,
then Australian Childhood Immunisation Register
ACIR Internet Helpline: 1300 650 039

Quarterly Outcomes
Payment & GPII Practice
Report (ACIR020A):

1800 246 101


Email: gpii@medicareaustralia.gov.au
Website: www.medicareaustralia.gov.au; click on Health
Care Providers, then Incentives & Allowances, then
General Practice Immunisation Incentive Scheme

State and Territory Government health authorities


Australian Capital Territory (02) 6205 2300
Immunisation Enquiry Line
New South Wales

Contact your local Public Health Unit, found


under Health in the White Pages

Northern Territory

(08) 8922 8044

Queensland

(07) 3234 1500

South Australia
Immunisation
Coordination Unit

(08) 8226 7177

Tasmania

(03) 6222 7724 or 1800 671 738

http://www.dh.sa.gov.au/pehs/

http://www.dhhs.tas.gov.au/publichealth/
immunisation/index.html
Victoria

1300 882 008


http://www.health.vic.gov.au/immunisation

Western Australia

(08) 9321 1312

332 The Australian Immunisation Handbook 9th Edition

Contact details for communicable disease control


Australian Capital Territory 24-hour Communicable Disease Control
Section: (02) 6205 2155
New South Wales

Contact your local Public Health Unit, found


under Health in the White Pages

Northern Territory

08301700 hrs: (08) 8922 8044 (After hours Royal Darwin


Hospital (08) 8922 8888 for CDC on-call doctor)

Queensland

Contact your local Public/Population Health Unit,


phone number listed in the White Pages

South Australia

24 hour general enquiries line: (08) 8226 7177

Tasmania

24 hour Hotline: 1800 671 738

Victoria

24 hour contact number: 1300 651 160

Western Australia

08301700hrs: (08) 9388 4999. After hours: (08) 9382 0553

Appendices

Appendix 1 333

Appendix 2: Handbook development


Introduction
The Handbook has been developed by the Australian Technical Advisory Group
on Immunisation (ATAGI), which provides advice to the Federal Minister for
Health and Ageing on the Immunise Australia Program and other related issues.
In addition to technical experts, ATAGIs membership includes a consumer
representative and general practitioners. ATAGI consulted with other expert
bodies, and with the National Health and Medical Research Council throughout
the development of the 9th edition of the Handbook. The Handbook does not
address the cost-effectiveness of different vaccines or different regimens;
since January 2006, the cost-effectiveness of vaccines has been assessed by
the Pharmaceutical Benefits Advisory Committee (PBAC), which advises
government on the funding of vaccines.1
The Handbook is designed as a general guide to inform clinicians on the safest
and most effective vaccination strategies, using the highest quality evidence
available in peer-reviewed literature, according to NHMRC guidelines.2-5 In
the absence of evidence at the highest level (well conducted randomised trials
and meta-analyses), recommendations were based on lower levels of evidence
such as case series. Where clinical guidelines were available on specific topics,
these were used to frame recommendations, if relevant, in the Australian
setting. Immunisation handbooks produced by comparable countries were also
consulted. If published sources were inadequate, recommendations were based
on expert opinion.
The draft of the 9th edition of the Handbook was available for public consultation
over a 6-week period from February to April 2007. The public comments received
were reviewed during an extraordinary meeting of ATAGI and, where necessary,
changes to the Handbook were incorporated.
The 9th edition of the Handbook is disseminated directly to all registered medical
practitioners. Additional hard copies are distributed to other immunisation
service providers via their State or Territory health authority. An electronic
version of the Handbook is accessible on the Immunise Australia Program website
http://immunise.health.gov.au/. Implementation of the recommendations as
stated in the Handbook is undertaken by immunisation providers in conjunction
with their State or Territory health authority, and the Immunise Australia
Program of the Australian Government Department of Health and Ageing.

Handbook literature search methodology


For each chapter, broad literature searches were conducted for theyears since the
last Handbook searches were performed using up to 18 databases, listed in Table
1. The purpose of these searches was to ensure that technical writers and chapter

334 The Australian Immunisation Handbook 9th Edition

sponsors had access to all relevant information from the latest medical literature
in identifying important issues relating to the updating of all Handbook chapters.
Table 1: Electronic databases searched
Electronic database

Time period

MEDLINE

20022006

PREMEDLINE (when required)

20022006

Cochrane Libraryincluding Cochrane Database of Systematic


Reviews, Database of Abstracts of Reviews of Effects, the Cochrane
Central Register of Controlled Trials (CENTRAL), the Health Technology
Assessment Database, the NHS Economic Evaluation Database

20022006

Cumulated Index Nursing & Allied Health


Literature (CINAHL) (when required)

20022006

EMBASE

20022006

Australian focused Informit databases (AMI, APA-FT, APAIS, APAIS


Health, ATSIhealth, Ausport Med, CINCH Health, DRUG,
Informit e-library, Health and Society, HIV A, Meditext, RURAL).

20022006

For specific questions arising in individual chapters, other databases and


additional resources such as Clinical Evidence were searched as necessary. The
scope and nature of the review differed for updating of existing chapters in the
Handbook and new chapters for the 9th edition.
Existing chapters in the Handbook

Various search methods were tested, including explode and focus options.
Exploded terms retrieve citations containing the term being searched and all the
narrower related terms in the database. Focus searches retrieve citations that
have the search term as the major focus of the item. In the trial searches, some
items of interest were missed using the focus method, thus exploded searches
were utilised. All subheadings assigned to the subject headings were generally
included.
In general, the search strategy consisted of the disease topic combined with the
terms immunisation/preventive health. Boolean operators AND, OR, and NOT
were used.

Appendix 2 335

Appendices

The medical and health literature was searched to identify relevant studies
and reviews regarding individual diseases and the vaccines available using the
electronic databases in Table 1. The search period was from 2002 to September
2006. The scope of the searches was broad, to ensure maximum retrieval and
minimise the exclusion of items of interest. Previous Handbook searches were
examined to determine the scope required for the new searches, and similar
search strategies were employed to ensure consistency of information retrieval.

To ensure relevant and accurate retrieval, thesaurus terms (the controlled


vocabulary terms used in the database) were used whenever possible. Keyword
searching was used only in the absence of an appropriate thesaurus term or if the
database did not have thesaurus terms. To facilitate relevant retrieval and to limit
what, in some instances, are very large search result sets, the following limits
were applied to the disease topic searches:
Publication year searches were generally limited to items published from
20022006, in order to retrieve items published since the searches completed
for the 8th edition of the Handbook.
Language searches were limited to items in English.
Human items discussing only animals were removed.
In vitro items discussing only in vitro studies were removed.
Abstracts search results restricted to items containing abstracts.
The search limits were slightly modified for some of the other searches. For
example, the Australian-specific searches did not have search results limited to
abstract only, to ensure that all Australian items were retrieved, including items
such as letters to the editor and editorials. In addition, the search result files were
edited to remove irrelevant items. Files were edited, where possible, to remove
duplication across the databases. There was often substantial duplication across
databases, as databases index some of the same journals.
More specific searches were undertaken when requested by chapter sponsors or
technical writers to provide additional information on a particular aspect of a topic.
New vaccine chapters
For the new vaccine chapters, Chapter 3.7 Human papillomavirus and Chapter
3.18 Rotavirus, search strategies employed for the existing chapters (as above)
were used. In addition, recommendations were formulated using a structured
clinical question using the PICO (Patient/population, Intervention, Comparison
and Outcome) format as per the NHMRC guidelines.3 Where possible, these
structured clinical questions informed both the searching and subsequent data
extraction processes. Search limits used for the new vaccine chapters differed
from those used for the existing chapters. To maximise retrieval and minimise
publication or other potential biases, no limitation by date, language or abstract
was applied. All papers identified as relevant from the PICO literature review
were critiqued using a standard proforma, according to whether they were
classified as studies of an intervention, diagnostic tests, prognosis, aetiology
or related to screening. Completion of a separate detailed data extraction form
was undertaken for each included study considered in the body of evidence for
each recommendation. These processes were completed in consultation with the
NHMRC-appointed Guideline Assessment Register consultant.

336 The Australian Immunisation Handbook 9th Edition

Complete details of the systematic literature review for the new vaccine chapters
in the Handbook may be found on the Immunise Australia website
www.immunise.health.gov.au.
The evidence presented in the included studies was assessed and classified
as described by the NHMRC.3 The consideration of important aspects of the
evidence supporting the intervention or recommendations included three main
domains:
strength of the evidence (level, quality and statistical precision),
size of the effect (including clinical importance), and
relevance of the evidence.
Grades of evidence were assigned to the recommendations in the new vaccine
chapters using the NHMRC considered judgment approach for assessing a body
of evidence (see Table 2). Grade A and B recommendations are generally based
on a body of evidence which can be trusted to guide clinical practice, whereas
Grade C and D recommendations must be applied carefully to individual clinical
and organisational circumstances and should be followed with care.
Table 2: Grades of recommendations4
Description

Body of evidence can be trusted to guide practice

Body of evidence can be trusted to guide practice in most situations

Body of evidence provides some support for recommendation(s)


but care should be taken in its application

Body of evidence is weak and recommendation


must be applied with caution

The level of evidence for the included studies for each recommendation is also
given, according to the NHMRC additional levels of evidence. Table 3 shows the
levels that apply to intervention studies (for other study types, such as aetiology,
prognosis or diagnosis, see http://www.nhmrc.gov.au/consult/index.htm). The
level of evidence indicates the study design used by the investigators to assess
the effectiveness of an intervention. The level assigned to a study reflects the
degree to which bias has been eliminated by the study design.3

Appendix 2 337

Appendices

Grade of
recommendation

Table 3: NHMRC Levels of evidence for intervention studies4


Intervention

Level of evidence

A systematic review of level II studies


A randomised controlled trial (RCT)

I
II

A pseudo-randomised controlled trial (eg. alternate


allocation or some other method)

III-1

A comparative study with concurrent controls:

III-2

Non-randomised, experimental trial


Cohort study
Case-control study
Interrupted time series with a control group
A comparative study without concurrent controls:

III-3

Historical control study


Two or more single arm study
Interrupted time series without a parallel control group
Case series with either post-test or pre-test/post-test outcomes

IV

Further information of how these grades were applied is given in the detailed
description of the systematic literature reviews for the new vaccine chapters, which
is available on the Immunise Australia website, www.immunise.health.gov.au.

Establishing Selected Dissemination Information (SDI) searches


Selected Dissemination Information (SDI) searches were established to enable
the ongoing collection of new relevant items on the search topics. This process
used the same search strategies as the previous searches. Search results are
automatically generated each time the databases (MEDLINE, EMBASE) are
updated, and a report is automatically sent to a nominated e-mail address. This
allowed inclusion of the most recent literature to be evaluated and referenced
throughout the revision and updating of the Handbook, where applicable.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

338 The Australian Immunisation Handbook 9th Edition

Appendix 3: Products registered in


Australia but not currently available
Products registered but not currently available
Arilvax

CSL Biotherapies/Novartis Vaccines (live attenuated


yellow fever virus (17D strain) vaccine)

Gamunex

Bayer Australia Pty Ltd (normal human immunoglobulin for IV use)

Pediacel

Sanofi Pasteur Pty Ltd (diphtheria-tetanus-acellular


pertussis-inactivated poliomyelitis vaccine-Hib)

Priorix-Tetra

GlaxoSmithKline (live measles-mumps-rubella-varicella vaccine)

ProQuad Frozen

CSL Biotherapies/Merck & Co Inc (live measlesmumps-rubella-varicella vaccine)

ProQuad

CSL Biotherapies/Merck & Co Inc (live measlesmumps-rubella-varicella vaccine)

Zostavax

CSL Biotherapies/Merck & Co Inc (live varicellazoster vaccine, frozen vaccine formulation)

Products previously available


CSL Biotherapies (diphtheria-tetanus vaccine, adult)

CDT

CSL Biotherapies (diphtheria-tetanus vaccine, child)

Diphtheria antitoxin

CSL Biotherapies

Diphtheria

CSL Biotherapies (vaccine, adult)

Diphtheria

CSL Biotherapies (vaccine, child)

Infanrix

GlaxoSmithKline (diphtheria-tetanusacellular pertussis vaccine, child)

Infanrix Hep B

GlaxoSmithKline (diphtheria-tetanus-acellular
pertussis-hepatitis B vaccine, child)

M-M-R II

CSL Biotherapies/Merck & Co Inc (measles-mumps-rubella vaccine)

Orochol

CSL Biotherapies (oral live recombinant


Vibrio cholerae CVD 103-HgR)

Quadracel

Sanofi Pasteur Pty Ltd (diphtheria-tetanus-acellular


pertussis-inactivated poliomyelitis vaccine, child)

Tet-Tox

CSL Biotherapies (tetanus vaccine)

Tripacel

Sanofi Pasteur Pty Ltd (diphtheria-tetanusacellular pertussis vaccine, child)

Typh-Vax (Oral)

CSL Biotherapies (live attenuated typhoid vaccine)

Tuberculin PPD

CSL Biotherapies (1000 units/mL)

Appendices

ADT

Appendix 3 339

Appendix 4: Components of vaccines used


in the National Immunisation Program*
For vaccines not listed in the National Immunisation Program, please refer
to individual product information leaflet as supplied with the vaccine, or the
Handbook chapter pertinent to that vaccine.
Vaccine component
(possible cause of
an allergy)

Vaccine brand

Antigen

Aluminium
hydroxide

Avaxim

Hepatitis A (HAV)

COMVAX

Haemophilus influenzae
typeb-hepatitis B (Hib-hepB)

Engerix-B

Hepatitis B (HBV) adult

Engerix-B

Hepatitis B (HBV) paediatric

Havrix Junior

Hepatitis A (HAV) paediatric

H-B-VAX II

Hepatitis B (HBV) adult

H-B-VAX II

Hepatitis B (HBV) paediatric

Infanrix-IPV

Diphtheria-tetanus-acellular pertussisinactivated poliomyelitis (DTPa-IPV)

Liquid PedvaxHIB

Haemophilus influenzae type b (Hib)

Menjugate Syringe

Serogroup C meningococcal conjugate

NeisVac-C

Serogroup C meningococcal conjugate

Vaqta

Hepatitis A (HAV) paediatric/adolescent

Boostrix

Diphtheria-tetanus-acellular
pertussis (dTpa) adult

GARDASIL

Human papillomavirus (HPV)

Infanrix hexa

Diphtheria-tetanus-acellular pertussis-hepatitis
B-inactivated poliomyelitis-Haemophilus
influenzae type b (DTPa-hepB-IPV-Hib)

Infanrix Penta

Diphtheria-tetanus-acellular pertussis-hepatitis
B-inactivated poliomyelitis (DTPa-hepB-IPV)

Adacel

Diphtheria-tetanus-acellular
pertussis (dTpa) adult

Meningitec

Serogroup C meningococcal conjugate

Prevenar

7-valent pneumococcal conjugate

COMVAX

Haemophilus influenzae
typeb-hepatitis B (Hib-hepB)

GARDASIL

Human papillomavirus (HPV)

Liquid PedvaxHIB

Haemophilus influenzae type b (Hib)

Vaqta

Hepatitis A (HAV) paediatric/adolescent

Aluminium
hydroxide/
phosphate

Aluminium
phosphate

Borax

340 The Australian Immunisation Handbook 9th Edition

Vaccine component
(possible cause of
an allergy)

Vaccine brand

Egg protein

All influenza
vaccines

Antigen

Influenza

Fluarix

Influenza

Fluvax

Influenza

Fluvirin

Influenza

Influvac

Influenza

Vaxigrip

Influenza

Adacel

Diphtheria-tetanus-acellular
pertussis (dTpa) adult

Avaxim

Hepatitis A (HAV)

Boostrix

Diphtheria-tetanus-acellular
pertussis (dTpa) adult

Fluad

Influenza

Fluarix

Influenza

Havrix Junior

Hepatitis A (HAV) paediatric

Infanrix hexa

Diphtheria-tetanus-acellular pertussis-hepatitis
B-inactivated poliomyelitis-Haemophilus
influenzae typeb (DTPa-hepB-IPV-Hib)

Infanrix-IPV

Diphtheria-tetanus-acellular pertussisinactivated poliomyelitis (DTPa-IPV)

Infanrix Penta

Diphtheria-tetanus-acellular pertussis-hepatitis
B-inactivated poliomyelitis (DTPa-hepB-IPV)

Vaqta

Hepatitis A (HAV) paediatric/adolescent

Vaxigrip

Influenza

Gelatin

Varivax Refrigerated

Varicella-zoster (VZV)

Gentamicin

Fluarix

Influenza

Formaldehyde

Influvac

Influenza

Kanamycin

Fluad

Influenza

Monosodium
glutamate (MSG)

Varivax Refrigerated

Varicella-zoster (VZV)

Appendix 4 341

Appendices

Fluad

Vaccine component
(possible cause of
an allergy)

Vaccine brand

Antigen

Neomycin

Avaxim

Hepatitis A (HAV)

Fluad

Influenza

Fluvax

Influenza

Fluvirin

Influenza

Havrix Junior

Hepatitis A (HAV) paediatric

Infanrix hexa

Diphtheria-tetanus-acellular pertussis-hepatitis
B-inactivated poliomyelitis-Haemophilus
influenzae typeb (DTPa-hepB-IPV-Hib)

Infanrix-IPV

Diphtheria-tetanus-acellular pertussisinactivated poliomyelitis (DTPa-IPV)

Infanrix Penta

Diphtheria-tetanus-acellular pertussis-hepatitis
B-inactivated poliomyelitis (DTPa-hepB-IPV)

Priorix

Measles-mumps-rubella (MMR)

Varilrix

Varicella-zoster (VZV)

Varivax Refrigerated

Varicella-zoster (VZV)

Vaxigrip

Influenza

Phenol

Pneumovax 23

23-valent pneumococcal polysaccharide

Phenoxyethanol

Adacel

Diphtheria-tetanus-acellular
pertussis (dTpa) adult

Avaxim

Hepatitis A (HAV)

Boostrix

Diphtheria-tetanus-acellular
pertussis (dTpa) adult

Havrix Junior

Hepatitis A (HAV) paediatric

Infanrix hexa

Diphtheria-tetanus-acellular pertussis-hepatitis
B-inactivated poliomyelitis-Haemophilus
influenzae typeb (DTPa-hepB-IPV-Hib)

Infanrix-IPV

Diphtheria-tetanus-acellular pertussisinactivated poliomyelitis (DTPa-IPV)

Infanrix Penta

Diphtheria-tetanus-acellular pertussis-hepatitis
B-inactivated poliomyelitis (DTPa-hepB-IPV)

Fluvax

Influenza

Fluvirin

Influenza

Infanrix hexa

Diphtheria-tetanus-acellular pertussis-hepatitis
B-inactivated poliomyelitis-Haemophilus
influenzae typeb (DTPa-hepB-IPV-Hib)

Infanrix-IPV

Diphtheria-tetanus-acellular pertussisinactivated poliomyelitis (DTPa-IPV)

Infanrix Penta

Diphtheria-tetanus-acellular pertussis-hepatitis
B-inactivated poliomyelitis (DTPa-hepB-IPV)

Polymyxin

342 The Australian Immunisation Handbook 9th Edition

Vaccine component
(possible cause of
an allergy)

Vaccine brand

Antigen

Thiomersal

Engerix-B

Hepatitis B (HBV) adult


(contains trace <2g/mL)

Engerix-B

Hepatitis B (HBV) paediatric


(contains trace <2g/mL)

Fluad

Influenza

Fluarix

Influenza

COMVAX

Haemophilus influenzae
typeb-hepatitis B (Hib-hepB)

Engerix-B

Hepatitis B (HBV) adult

Engerix-B

Hepatitis B (HBV) paediatric

GARDASIL

Human papillomavirus (HPV)

H-B-VAX II

Hepatitis B (HBV) adult

H-B-VAX II

Hepatitis B (HBV) paediatric

Infanrix hexa

Diphtheria-tetanus-acellular pertussis-hepatitis
B-inactivated poliomyelitis-Haemophilus
influenzae typeb (DTPa-hepB-IPV-Hib)

Infanrix Penta

Diphtheria-tetanus-acellular pertussis-hepatitis
B-inactivated poliomyelitis (DTPa-hepB-IPV)

Yeast

* Please note that vaccine manufacture is subject to ongoing refinement and change.
Therefore, the following information may change. This information was current at the time
of preparation.
This table has been adapted with permission from Dr T. McGuire.1

Please also refer to Appendix 5, Commonly asked questions about vaccination for more specific
information about these various constituents.

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

Appendix 4 343

Appendices

If the person to be vaccinated has had an anaphylactic reaction to any of the vaccine
components, administration of that vaccine is contraindicated.

Appendix 5: Commonly asked


questions about vaccination
This chapter contains information for providers to refer to when responding
to questions and concerns about immunisation. It covers general questions on
adult and childhood vaccination, including contraindications and precautions.
In addition, a discussion on some of the more recent concerns about vaccination
is included, covering issues relating to vaccine safety, vaccine content,
immunisation as a possible cause of some illnesses of uncertain origin, and the
need for vaccination.
This appendix is divided into 4 sections:

1. General questions

2. Contraindication and precautions

3. Responding to questions and concerns about immunisation

4. Where can I get more information about vaccination?

1. General questions
(i) How does vaccination work?
When a healthy person becomes infected with a virus, eg. measles, the body
recognises the virus as an invader, produces antibodies which eventually destroy
the virus and recovery occurs. If contact with the measles virus occurs again
in the future, the bodys immune system remembers the measles virus and
produces an increase in antibodies to destroy the virus.
Vaccination is the process that is used to stimulate the bodys immune system in
the same way as the real disease would, but without causing the symptoms of
the disease. Most vaccines provide the body with memory so that an individual
doesnt get the disease if exposed to it.
Vaccination conveys immunity to diseases by a process called active immunity,
which can be achieved by administration of either inactivated (ie. not live) or
live attenuated organisms or their products. Live vaccines are attenuated, or
weakened, by growing the organism through serial culturing (or passaging)
steps in various tissue culture media. Inactivation is usually done using heat
or formalin (sometimes both). Inactivated vaccines may include the whole
organism (such as oral cholera vaccine), the toxin produced by the organism
(such as tetanus and diphtheria vaccines), or specific antigens (such as Hib and
pneumococcal vaccines). In some cases, the antigen is conjugated (ie. chemically
linked) with proteins to facilitate the immune response. Inactivated viral vaccines
may include whole viruses (such as IPV and hepatitis A vaccines) or specific
antigens (such as influenza and hepatitis B vaccines). Live attenuated viral
vaccines include MMR, rubella, varicella and yellow fever vaccines.

344 The Australian Immunisation Handbook 9th Edition

Immunity can also be acquired passively by the administration of


immunoglobulins. Such immunity is immediate and is dose-related and
transient. For example, measles or hepatitis B immunoglobulin can be used
promptly after exposure in an unimmunised person to help reduce the chance of
catching measles or hepatitis B from the exposure.

(ii) What is the correct site for vaccination of children?


The top, outer part of the thigh (the vastus lateralis muscle) is the recommended
site for injections for infants <12months of age. The deltoid region of the upper
arm is the recommended site for vaccination of children 12months of age
because it is associated with fewer local reactions and has sufficient muscle bulk
to facilitate the injection. However, the vastus lateralis muscle can also be used.
The ventrogluteal area is an alternative site. (See Section 1.4.6, Recommended
injection sites and Section 1.4.8, Identifying the injection site).
Rotavirus vaccines are administered by the oral route and must never be injected.

(iii) How many injections can be given into the


same limb in a child aged <12months?

(iv) When should preterm infants be vaccinated?


Babies born at <32weeks gestation or <2000 g birth weight should receive
their first dose of hepatitis B vaccine either at birth (within the first fewdays of
life) or at 2months of age. Immunisation schedules differ in different areas of
Australia according to which combination vaccines are routinely used in that
State or Territory. The routine 2-month vaccines containing the antigens DTPaIPV-Hib-hepB, 7vPCV and rotavirus should be given 2months after birth as
normal, unless an infant is very unwell. Very unwell can be interpreted in many
ways but, in general, reflects that the premature neonate is particularly unstable.
Delaying the 2-month vaccines is rarely required. If any preterm infant has the
2-month vaccines delayed, it should be remembered that the infant doses can
be given 1month apart rather than 2months. Hence, if an infant receives the
2-month vaccines at 3months then the 4month vaccines should still be given at
4months of age.

Appendix 5 345

Appendices

More than 1 vaccine can be safely given into a limb at the same immunisation
visit. Most States and Territories have routine immunisation schedules that
include 3 injections during the primary course for children <12months of
age. In this case, 2 injections can be given into the same leg into the vastus
lateralis muscle and the third injection in the other vastus lateralis muscle (or
an alternative is the ventrogluteal site). The injections should be given at least
25mm (2.5 cm) apart. Use separate sterile injection equipment for each vaccine
administered. The accompanying documentation should indicate clearly which
vaccines were given into which site (eg. left leg upper/left leg lower).

When Liquid PedvaxHIB is used in an extremely preterm baby (<28weeks


gestation or <1500 g birth weight), an additional dose should be given at
6months of age.
Further explanation of the special immunisation needs of premature babies is
provided in Section2.3.2 Vaccination of women planning pregnancy, pregnant or
breastfeeding women, and preterm infants.

(v) Do elderly people (>65years) who have no chronic


illnesses need the influenza vaccine?
Yes. Age is an independent risk factor for influenza. Vaccination of those aged
>65years, regardless of the presence or absence of chronic illness, reduces
mortality by up to 50% in the winter period in this age group (see Chapter 3.9,
Influenza).1,2 The healthy elderly should also receive the 23-valent pneumococcal
polysaccharide vaccine (see Chapter 3.15, Pneumococcal disease).

(vi) Should adults receive pertussis (whooping cough) vaccine boosters?


Yes. Two brands of acellular pertussis vaccines, both combined with tetanus and
diphtheria antigens, are now available for adolescents and adults. dTpa vaccines
are recommended in Australia for booster vaccination of individuals 8years of
age who have previously had a primary course of diphtheria-tetanus-pertussis
vaccine. dTpa vaccines have a lower content of diphtheria and pertussis antigens
than DTPa formulations for young children.
A recent study showed that adults can be protected against pertussis after a
single dose of dTpa. No recommendations about the need for further boosters
using adolescent/adult formulation dTpa have been made at this time.
A single dose of dTpa is recommended for the following groups (unless
contraindicated or they have already received a previous dose of dTpa):
adults working with young children. Vaccination is especially recommended
for childcare workers;
all healthcare workers;
adults planning a pregnancy, or for both parents as soon as possible
after delivery of an infant (preferably before hospital discharge), unless
contraindicated.3 Other adult household members, grandparents and carers
of young children should also be vaccinated. This recommendation is based
on evidence from several studies of infant pertussis cases, which indicated
that family members, particularly parents, were identified as the source of
infection in more than 50% of cases and were the presumed source in a higher
proportion;
any adult expressing an interest in receiving a booster dose of dTpa.
Adults 50years of age who have not previously received dTpa vaccine should
also be offered vaccination. See Chapter 3.3, Diphtheria, Chapter 3.14, Pertussis or
Chapter 3.21, Tetanus in this Handbook.

346 The Australian Immunisation Handbook 9th Edition

Contraindications to adolescent/adult formulation dTpa are discussed in


Chapter 3.3, Diphtheria, Chapter 3.14, Pertussis and Chapter 3.21, Tetanus in this
Handbook and include previous anaphylactic reaction to any vaccine component.
If the patient has never received a primary course of dT, see Chapter 3.3,
Diphtheria, Chapter 3.14, Pertussis or Chapter 3.21, Tetanus in this Handbook.

(vii) A parent wants a child to receive his/her vaccines


separately. Why cant they do this?
There is no scientific evidence or data to suggest that there are any benefits in
receiving vaccines such as MMR or DTPa as separate monovalent vaccines.
Using the example of MMR vaccine, there is no individual mumps or measles
vaccine licensed for use in Australia. If these vaccines were to be administered
individually it would require 3 separate vaccines which would unnecessarily
increase discomfort for the child. In addition, if these monovalent vaccines were
not given on the same day, they would need to be spaced 1month or more apart
which would increase the risk of that child being exposed to serious vaccinepreventable diseases. A policy of providing separate vaccines would cause some
children to not receive the entire course, and combination vaccines can offer a
reduced amount of vaccine stabiliser and adjuvant compared to 3 individual
vaccine doses.

(viii) Is vaccination compulsory? What happens


if children do not get vaccinated?
Vaccination is not compulsory in Australia.

If a parent decides not to have a child vaccinated and, if cases of certain vaccinepreventable diseases occur at that childs day-care centre or school, the parent
may, in some circumstances, be required to keep the unvaccinated child at home
until the incubation period for that particular disease has passed or no further
cases have occurred in that setting.

2. Contraindications and precautions


If you have any concerns about whether to proceed with vaccination, please
seek expert advice. See Appendix 1, Contact details for Australian, State and
Territory Government health authorities and communicable disease control.

(i) What are the absolute contraindications to childhood vaccination?


True contraindications to the childhood vaccines are extremely rare (see relevant
chapters), and include only anaphylaxis to any of the particular vaccines
components, and anaphylaxis following a previous dose of that vaccine.

Appendix 5 347

Appendices

The Maternity Immunisation Allowance and Child Care Benefit are parent
incentive payments that are paid where a child is up-to-date with his/
her immunisations or the parent has obtained an appropriate medical or
philosophical exemption.

NB. Anaphylaxis following ingestion of eggs does not contraindicate MMR


vaccine, as the vaccine viruses are not grown in eggs and the vaccine does not
contain any egg protein4 (see Chapter 3.11, Measles).

(ii) What are the contraindications to further doses


of pertussis-containing vaccines?
Further doses of DTPa are contraindicated in those who have had a previous
immediate severe allergic or anaphylactic reaction to vaccination with DTPa.
A previous simple febrile convulsion or pre-existing neurological disease is not a
contraindication to pertussis-containing vaccines.

(iii) What are the precautions to childhood vaccination?


In general, children with impaired immunity or on immunosuppressive therapy
should not be given live vaccines (see (vii) to (ix) below).

(iv) Should a child with an intercurrent illness be vaccinated?


A child with a minor illness (without systemic illness and with a temperature
<38.5C) may be safely vaccinated. Infants and children with minor coughs and
colds without fever, or those receiving antibiotics in the recovery phase of an
acute illness, can be vaccinated safely and effectively. In a child with a major
illness or high fever 38.5C, vaccination should be postponed until the child is
well. If vaccination were to be carried out during such an illness, the fever might
be confused with vaccine side effects and might also increase discomfort to the
child. In such cases, it is advisable to defer vaccination and arrange for the child
to return for vaccination when well again.

(v) Should children with epilepsy be vaccinated?


Yes. Stable neurological disease (such as epilepsy) is not a reason to avoid giving
vaccines like pertussis (whooping cough). Children who are prone to fits should
have paracetamol before and for 48 hours after vaccination to reduce the chance
of a fever after vaccination bringing on a convulsion. A family history of fits or
epilepsy is not a reason to avoid vaccination.

(vi) Should children with neurological disease receive


the normal vaccination schedule?
Yes. Children with neurological disease are often at increased risk of
complications from diseases like measles, influenza and whooping cough, as they
can be more prone to respiratory infections and chest problems. It is important
that these children be immunised, on time, as recommended in the National
Immunisation Program schedule.

(vii) Are steroids a contraindication to vaccination?


Live vaccines such as MMR, BCG and varicella-zoster vaccines, should
not be given to children or adults receiving high dose oral or parenteral
corticosteroid therapy for more than 2weeks. High dose oral corticosteroid

348 The Australian Immunisation Handbook 9th Edition

therapy is defined as more than 2mg/kg per day prednisolone for more than
1week. This is because steroids, in large doses, greatly suppress the immune
system which means that, not only is the vaccine unlikely to be effective, but
there is an increased chance of an adverse event occurring as a result of the
immunosuppression.
Inactivated vaccines, eg. DTPa-hepB-IPV, may be less effective in this group but
are not contraindicated. Therapy with inhaled steroids is not a contraindication
to vaccination.

(viii) Should vaccines be given to children who have


problems with their immune systems?
Children with impaired immunity or those on immunosuppressive therapy should
not be given live viral vaccines such as MMR, varicella, and rotavirus vaccines.5
HIV-infected children may be given MMR vaccine provided they do not have
severely impaired immunity (see Table 2.3.4 Immunological categories based on
age-specific CD4 counts and percentage of total lymphocytes). The contacts of children
with impaired immunity can be given MMR without any risk of transmission.
The rash seen in a small percentage of MMR vaccine recipients, usually
betweendays 5 to 12 post vaccination, is not infectious.

Live viral vaccines can be given to children with leukaemia and other
malignancies who have been on chemotherapy at least 3months after they have
completed chemotherapy, provided there are no concerns about their immune
status. Such measures would normally be carried out under the supervision of
the childs oncologist (see Section 2.3.3, Vaccination of individuals with impaired
immunity due to disease or treatment).

(ix) What vaccines should children with HIV infection receive?


Children with HIV (human immunodeficiency virus) infection should have all
routine inactivated vaccines on the National Immunisation Program schedule.
Varicella vaccine is generally contraindicated in children with HIV, as it can cause
disseminated varicella infection. However, it may be considered for asymptomatic
or mildly symptomatic HIV-infected children, after weighing up the potential
risks and benefits. This should be discussed with the childs specialist.

Appendix 5 349

Appendices

It is highly recommended that non-immune household contacts of children with


impaired immunity receive varicella vaccine. There is an almost negligible risk
of transmitting varicella vaccine virus from a vaccine-related vesicular rash to
contacts. However, vaccine-related rash occurs in 3 to 5% of vaccinees either
locally at the injection site or generalised, with a median of only 25 lesions.
This small infection risk of the less virulent attenuated vaccine strain is far
outweighed by the high risk of non-immune contacts catching wild varicella
infection and transmitting the virus to the household member with impaired
immunity via respiratory droplets or from the large number of skin lesions that
occur with wild varicella infection (a median of 300 to 500 lesions).

MMR vaccine can be given to children with HIV, depending on their CD4 counts
(see point (viii) above). Children with HIV infection should also be vaccinated
against pneumococcal disease (see Chapter 3.15, Pneumococcal disease). Influenza
vaccine is also recommended for HIV-infected children. They should not be given
BCG, due to the risk of disseminated infection (see Section 2.3.3, Vaccination of
individuals with impaired immunity due to disease or treatment).

(x) Should chronically ill children be vaccinated?


In general, children with chronic diseases should be vaccinated as a matter of
priority because they are often more at risk from complications from vaccinepreventable diseases. Annual influenza vaccine is highly recommended for
chronically ill children and their household contacts.
Care is needed with the use of live attenuated viral vaccines in situations where
the childs illness, or its treatment, may result in impaired immunity. Advice
may need to be sought on these patients to clarify the safety of live viral vaccine
doses.

(xi) Should children be vaccinated while the childs mother is pregnant?


There is no problem with giving routine vaccinations to a child whose mother
is pregnant. MMR vaccine viruses are not transmissible. Administration of
varicella vaccine to household contacts of non-immune pregnant women is safe.
Transmission of varicella vaccine virus is very rare. There is an almost negligible
risk of transmitting varicella vaccine virus from a vaccine-related vesicular rash
to contacts. However, vaccine-related rash occurs in 3 to 5% of vaccinees either
locally at the injection site or generalised, with a median of only 25 lesions.
Furthermore, vaccinating the child of a pregnant mother will reduce the risk of
her being infected by her offspring with the more virulent wild virus strain if she
is not immune.

(xii) Should children with allergies be vaccinated? What precautions


are required for atopic or egg-sensitive children?
Asthma, eczema and hay fever are not contraindications to any vaccine on the
childhood schedule unless the child is receiving high-dose steroid treatment (see
point (vii) above). For other allergies, see Appendix 4, Components of vaccines
used in the National Immunisation Program, and the relevant vaccine product
information (PI) enclosed in the vaccine package. Unless the child (or person
being vaccinated) has an allergy to a specific constituent of a vaccine (or has
another contraindication) there is no reason not to vaccinate.
An important exception is anaphylactic sensitivity to eggs, characterised by
generalised hives, swelling of the mouth or throat, difficulty breathing, wheeze,
low blood pressure, and shock. If a person has a history of severe egg allergy,
influenza, yellow fever and Q fever vaccines should not be given. Because MMR
vaccine viruses are not cultured in eggs and the vaccine does not contain egg
protein, MMR can be given safely to those with anaphylactic sensitivity to eggs.4

350 The Australian Immunisation Handbook 9th Edition

Simple dislike of eggs or having diarrhoea or stomach pains after eating eggs
are not reasons to avoid MMR and these children require no special precautions.
These children can also have all other routine vaccines without special
precautions.4
Families with questions about allergies and vaccines are encouraged to discuss this with
their immunisation service provider to have any questions promptly answered to avoid
unnecessary delays of vaccine doses.

3. Responding to questions and concerns about immunisation


Some people express concerns about immunisation. These mostly relate to
whether the vaccine is safe and whether vaccines weaken the immune system
of the child. Providers should listen to and acknowledge peoples concerns.
Providers should discuss the risks and benefits of immunisation with parents/
carers honestly and in a non-defensive manner. Parents/carers and adult
vaccine recipients should receive accurate information on the risks from vaccinepreventable diseases and information about vaccine side effects and adverse
events (see table inside the front cover Comparison of the effects of diseases and the
side effects of vaccines). The following section responds to some concerns raised
about the safety of immunisation, and examines the scientific evidence in order
to assist providers and parents in making an informed choice about the risks and
benefits of vaccination.

a) Vaccine safety
(i) How safe are vaccines?

After introduction into vaccination schedules, there is continuing surveillance of


efficacy and safety through trials and post-marketing surveillance. In Australia,
there are regional and national surveillance systems actively seeking any adverse
events following immunisation. This is necessary, as sometimes problems occur
after vaccines are registered for use. Regular Australian surveillance reports are
published in the journal Communicable Diseases Intelligence.6

(ii) Can too many vaccines overload or suppress the natural immune system?
No. The increase in the number of vaccines and vaccine doses given to children
has led to concerns about the possible adverse effects of the aggregate vaccine
exposure, especially on the developing immune system. In day-to-day life, all
children and adults confront enormous numbers of antigens and the immune
system responds to each of these in various ways to protect the body. Studies of

Appendix 5 351

Appendices

Before vaccines are made available for general use they are tested for safety and
efficacy in clinical trials and then in large trials, otherwise known as Phase 2 and
3 trials. All vaccines marketed in Australia are manufactured according to strict
safety guidelines and are evaluated by the Therapeutic Goods Administration
to ensure they are efficacious and are of adequate quality and safety before
marketing approval is granted.

the diversity of antigen receptors indicate that the immune system can respond
to an extremely large number of antigens. In addition, the number of antigens
received by children during routine childhood vaccination has actually decreased
compared with several decades ago. This has occurred in spite of the increase in
the total number of vaccines given, and can be accounted for by the removal of 2
vaccines smallpox vaccine (which contained about 200 different proteins), and
whole-cell pertussis vaccine (about 3000 distinct antigenic components) from
routine vaccination schedules. In comparison, the acellular pertussis vaccine
currently used in Australia has only 3 to 5 pertussis antigens.7

(iii) Do vaccines cause disease?


Some studies have suggested a temporal link between vaccinations and
certain medical conditions, such as asthma, multiple sclerosis, and diabetes.
The allegations of a link are often made for a disease of unknown cause. The
appearance of a certain medical condition after vaccination does not necessarily
imply that they are causally related. Importantly, however, once an issue is raised
it needs prompt research, discussion and then education to avoid propagating a
myth. In many cases, subsequent epidemiological studies have indicated that the
association is due to chance alone. The following is a list of concerns that have
been raised.
Does MMR vaccine cause inflammatory bowel disease or autistic spectrum
disorder?8
In 1998, Wakefield et al (Royal Free Hospital, London)9 published a case series
study with 12 children suggesting that MMR vaccine caused inflammatory bowel
disease (IBD), which then resulted in decreased absorption of essential vitamins
and nutrients through the intestinal tract. They proposed that this could result in
developmental disorders such as autism.
Following the studys publication, Wakefield suggested that it may be 3 live
viruses in the 1 vaccine which was causing the development of the subsequent
disorders. He suggested it was preferable to provide MMR vaccination as 3
separate vaccines, a suggestion with no supportive evidence.9
This study had several weaknesses. First, finding out whether or not MMR
causes autism is best determined by comparing the incidence of autism in
vaccinated versus unvaccinated children. However, the researcher included
only vaccinated children. Second, the author claimed that gastrointestinal
inflammation contributes to autism. However, in several of the children,
their behavioural problems appeared before the onset of bowel disease.10,11
Furthermore, the study was primarily based on parental recall of when the bowel
disease and developmental disorders first appeared, and people are more likely
to have linked changes in behaviour with memorable events such as vaccination.
The Royal Free Hospital study was conducted on a very selective group of
patients, all referred to the hospital for gastrointestinal ailments, and such a case
series analysis is unable to determine causal links.

352 The Australian Immunisation Handbook 9th Edition

The onset of autism and MMR vaccination may coincide because the average age
at which parents report concerns about child development is 18 to 19months,
and more than 90% of children in the UK receive MMR vaccine before their 2nd
birthday.12 More rigorous and larger epidemiological studies have found no
evidence of an association.13-16
A review by the World Health Organization concluded that current scientific data
do not permit a causal link to be drawn between the measles virus and autism
or IBD.17An extensive review published in 2004 by the Institute of Medicine,
an independent expert body in the United States, concluded that there is no
association between the MMR vaccine and the development of autism.18 Reviews
by the American Academy of Pediatrics, The British Chief Medical Officer, the
UK Medical Research Council, Canadian experts, and numerous other scientific
experts have stated that there is no link between autism or IBD and the measles
vaccine.14,19-21
See Chapter 3.11, Measles for further information. There is also an
MMR vaccine decision aid designed for parents available at
http://www.ncirs.usyd.edu.au/decisionaid/index.html.
Do childhood immunisations cause asthma?
There is no evidence that vaccination causes or worsens asthma. It is especially
important that children with asthma be vaccinated like other children, as
catching a disease like whooping cough can make an asthma attack worse.
Although influenza vaccine is not routinely recommended for all asthmatics,
it is recommended for severe asthmatics, such as those requiring frequent
hospitalisation.22
There is no reliable evidence that the hepatitis B vaccine causes multiple sclerosis
(MS). With millions of hepatitis B vaccinations administered worldwide, it is
likely that surveillance systems in some countries will receive some reports of
MS, which seem to be related in time to vaccinations. As with all such reports,
however, they suggest only the possibility of an association. Subsequent studies
have found no increase in incidence of MS, or even relapse of MS, after hepatitis
B vaccination.23-27
In response to a single study by Hernn et al,28 the World Health Organization
Global Advisory Committee on Vaccine Safety released the following statement:
multiple studies and review panels have concluded that there is no link between
MS and hepatitis B vaccination.29
In addition, a review by the Institute of Medicine Immunization Safety Review
Committee in 2003 found no link between hepatitis B vaccine and certain
neurological disorders such as MS.30 A systematic review from the Cochrane
Vaccines Field in 2003 also found no evidence of an association between hepatitis
B vaccine and MS.31 Recent statements by the World Health Organization and the
US Centers for Disease Control and Prevention support this position.29,32,33

Appendix 5 353

Appendices

Does hepatitis B vaccine cause multiple sclerosis?

Do some vaccines cause Mad Cow Disease?


Variant Creutzfeldt-Jakob disease (vCJD) is considered to be the human
equivalent of bovine spongiform encephalopathy (BSE, also known as mad
cow disease). There is no evidence that any case of vCJD has resulted from the
administration of any vaccine product, despite millions of doses of vaccine being
administered worldwide. Concerns about the risk of transmission of this disease
arose because the production of some vaccines requires bovine derivatives such
as fetal bovine serum. In Australia, the Therapeutic Goods Administration has
confirmed that the vaccines available in this country contain bovine materials
preferentially sourced from BSE-free areas, and that they undergo appropriate
purification treatment. Therefore, although some vaccines carry a theoretical risk
of transmissible spongiform encephalopathies, this risk is infinitesimally small
(estimated at less than 1 in a billion).34 The benefits of vaccination are considered
to far outweigh any theoretical risk of BSE transmission.35
Is there a link between vaccination and Sudden Infant Death Syndrome
(SIDS)?
Despite extensive studies, there is no evidence that vaccination causes SIDS (cot
death). Deaths do occasionally occur shortly after vaccination but the relationship
is a chance association, since SIDS tends to happen in babies of 26months of
age, whether they are vaccinated or not.36 Many studies have conclusively shown
that SIDS is not caused by immunisation. In addition, some studies have found a
lower rate of SIDS in immunised children.37-39
Does immunisation cause diabetes?
In 1997, a study from Finland suggested a link between Hib vaccination and type
1 diabetes.40 However, subsequent reanalysis of the data did not support such a
link.41,42 The conclusion that there is no causal link between any of the childhood
vaccines and diabetes has also been supported by a subsequent review of the
literature, and the conclusions of 2 workshops held in the USA in 1998.8,41-43
Does influenza vaccine cause flu?
No. It is not possible for influenza vaccine to cause flu as it is not a live viral
vaccine. As some people experience side effects such as a mild fever after the
vaccine, it is understandable that they may confuse these symptoms with
actually having the flu. In addition, the influenza vaccine is recommended to
be given at the commencement of the flu season. Hence, it is possible that a
person who has contracted, and is incubating, influenza during vaccination
will mistakenly believe the vaccine to be causal. In addition, influenza vaccine
is given at the very time of year when there are a lot of upper respiratory tract
infections (URTIs) around. It is not uncommon for someone to attribute an URTI
within aweek of an influenza vaccine to the vaccine dose. Importantly, URTI
symptoms occurring after influenza vaccine should not put people off having the
vaccine the following year.

354 The Australian Immunisation Handbook 9th Edition

b) Vaccine content44
(See also Appendix 4 for a list of vaccines used in the NIP which contain these
compounds and refer to the relevant vaccine product information (PI) enclosed
in the vaccine package.)
Preservatives
Preservatives are used to prevent fungal and or bacterial contamination of the
vaccine. They include thiomersal, phenoxyethanol, and phenol.

(i) Thiomersal
Thiomersal (or thimerosal) is a compound which is partly composed of mercury,
ethylmercury. It has been used in very small amounts in vaccines for about
60years, to prevent bacterial and fungal contamination of vaccines. In the past,
the small amount of thiomersal in vaccines was one of several potential sources
of mercury. Diet (such as some seafood) and other environmental sources are also
possible sources of mercury. Vaccines used in the past, such as DTP, contained
only 25g of thiomersal per dose.

Currently, all vaccines on the NIP for children <5years of age are now either free
of thiomersal, or contain a reduced (trace) amount of thiomersal.
There are certain vaccines that are still most effectively manufactured using a
trace amount of thiomersal as the preservative, eg. influenza vaccine. Infants
from 6months of age can be given influenza vaccine safely.

(ii) Phenoxyethanol
2-Phenoxyethanol is an aromatic ether alcohol used as a preservative in many
vaccines. It is also used as a preservative in cosmetics. 2-Phenoxyethanol is used
in vaccines as an alternative preservative to thiomersal.

Appendix 5 355

Appendices

Mercury causes a toxic effect after it reaches a certain level in the body. Whether
or not it reaches a toxic level depends on the amount of mercury consumed
and the persons body weight; individuals with very low body weight are
usually more susceptible to toxic effects from a certain intake of mercury.
Thus, the possibility existed that vaccination of newborn babies, particularly
those of very low birth weight, with repeated doses of thiomersal-containing
vaccines, might have resulted in levels of mercury above the recommended
guidelines. Thiomersal was removed from vaccines in response to the above
theoretical concern and to reduce total exposure to mercury in babies and young
children in a world where other environmental sources may be more difficult to
eliminate.45-47

(iii) Phenol
Phenol is an aromatic alcohol used as a preservative in a few vaccines.
Adjuvants
Adjuvants are compounds used to enhance the immune response to vaccination
and include various aluminium salts such as aluminium hydroxide, aluminium
phosphate and potassium aluminium sulphate (alum). A recent review of all
available studies of aluminium-containing diphtheria, tetanus and pertussis
vaccines (either alone or in combination) found no evidence that aluminium salts
in vaccines cause any serious or long-term adverse events.48

Aluminium
A small amount of aluminium salts has been added to some vaccines for
about 60years. Aluminium acts as an adjuvant, which improves the protective
response to vaccination by keeping antigens near the injection site so they can
be readily accessed by cells responsible for inducing an immune response. The
use of aluminium in vaccines means that, for a given immune response, less
antigen is needed per dose of vaccine, and a lower number of total doses are
required. Although aluminium-containing vaccines have been associated with
local reactions and, less often, with the development of subcutaneous nodules at
the injection site, other studies have reported fewer reactions with aluminiumadsorbed vaccines than with unadsorbed vaccines. Concerns about the longerterm effects of aluminium in vaccines arose after some studies suggested a link
between aluminium in the water supply and Alzheimers disease, but this link
has never been substantiated. The amount of aluminium in vaccines is very
small and the intake from vaccines is far less than that received from diet or
medications such as some antacids.49,50
Additives
Additives are used to stabilise vaccines in adverse conditions (temperature
extremes of heat and freeze drying) and to prevent the vaccine components
adhering to the side of the vial.
Examples of additives include:
lactose and sucrose (both sugars);
glycine and monosodium glutamate or MSG (both are amino acids or salts of
amino acids);
gelatin, which is partially hydrolysed collagen usually of bovine or porcine
origin. Some members of the Islamic and Jewish faiths may object to
vaccination, arguing that vaccines can contain pork products. Scholars of
the Islamic Organization for Medical Sciences have determined that the
transformation of pork products into gelatin will sufficiently alter them
thus making it permissible for observant Muslims to receive vaccines, even
if the vaccines contain porcine gelatin. Likewise, leaders of the Jewish faith

356 The Australian Immunisation Handbook 9th Edition

have also indicated that pork-derived additives to medicines are permitted.


Further information may be obtained from the following websites
http://vaccinesafety.edu/Porcine-vaccineapproval.htm and
http://www.immunize.org/concerns/porcine.pdf;
human serum albumin (protein).
Manufacturing residuals
Manufacturing residuals are residual quantities of reagents used in the
manufacturing process of individual vaccines. They include antibiotics (such
as neomycin or polymyxin), inactivating agents (eg. formaldehyde) as well
as cellular residuals (egg and yeast proteins), traces of which may be present
in the final vaccine. Antibiotics are used during the manufacturing process to
ensure that bacterial contamination does not occur; traces of these antibiotics
may remain in the final vaccine. Inactivating agents are used to ensure that
the bacterial toxin or viral components of the vaccine are not harmful, but will
result in an immune response. Cellular residuals are minimised by extensive
filtering. However, trace amounts may be present in the final product. The most
commonly found residual is formaldehyde.

Formaldehyde

Other
Vaccines also may be made up in sterile water or sterile saline (salt-water).

(c) The need for immunisation


(i) Isnt natural immunity better than immunity from vaccination?
While vaccine-induced immunity may diminish with time without boosters
(vaccine or contact with wild-type infection), natural immunity, acquired by
catching the disease, is usually life-long, with the exception of pertussis. The
problem is that the wild or natural disease has a higher risk of serious illness
and occasionally death. Children or adults can be revaccinated (with some but
not all vaccines) if their immunity from the vaccines falls to a low level or if
previous research has shown that a booster vaccination is required for long-term
protection. It is important to remember that vaccines are many times safer than
the diseases they prevent.

Appendix 5 357

Appendices

Formaldehyde is used during the manufacture of many vaccines. For example,


with tetanus vaccines, formaldehyde is used to detoxify the tetanus toxin protein
produced. The non-toxic protein which becomes the active ingredient of the
vaccine is further purified to remove contaminants and any excess (unreacted or
unbound) formaldehyde. The current standard applicable to vaccines for human
use in Australia is less than 0.02% w/v of free formaldehyde. The maximum
amount of free formaldehyde detected by the Therapeutic Goods Administration
during testing of vaccines registered in Australia has been 0.004% w/v, which is
well below the standard limit.

(ii) Diseases like measles, polio, whooping cough and diphtheria


have already disappeared from most parts of Australia. Why do we
need to keep vaccinating children against these diseases?
Although these diseases are much less common now, they still exist. The
potential problem of disease escalation is kept in check by routine vaccination
programs. In countries where vaccination rates have declined, vaccinepreventable diseases have sometimes reappeared. For example, Holland has one
of the highest rates of fully vaccinated people in the world. However, in the early
1990s, there was a large outbreak of polio among a group of Dutch people who
belonged to a religious group that objected to vaccination. While many of these
people suffered severe complications like paralysis, polio did not spread into the
rest of the Dutch community. This was due to the high rate of vaccination against
polio, which protected the rest of the Dutch community.
There have been recent outbreaks of whooping cough, measles and rubella in
Australia, and a number of children have died. Cases of tetanus and diphtheria,
although rare, still occur. Thus, even though these diseases are much less common
now than in the past, it is necessary to continue to protect Australian children, so
that the diseases cannot re-emerge to cause large epidemics and deaths.
Also, many of the diseases which we vaccinate our children against are still
common in other areas of the world. For example, measles still occurs in many
Asian countries and many people take holidays or travel for business to these
areas. Therefore, it is possible for non-immune individuals to acquire measles
overseas, and with the speed of air travel, arrive home and be able to pass
measles onto those around them if they are unprotected. Measles is highly
infectious and can infect others for several hours after an infected person has left
a room. Vaccination, while not 100% effective, can minimise a persons chance
of catching a disease. The more people who are vaccinated the less chance of a
disease, such as measles, spreading widely in the community. This is referred to
as herd immunity.

(iii) Why do some children get the disease despite being vaccinated?
This is possible, since no vaccine is 100% effective. A small proportion of those
who are vaccinated will remain susceptible to the disease. However, in the cases
in which illness does occur in vaccinated individuals, the illness is usually much
less severe than in those who were not vaccinated. The protection provided by
the same vaccine to different individuals can differ. For example, if 100 children
are vaccinated with MMR, 5 to 10 of the fully vaccinated children might still
catch measles, mumps or rubella (although the disease will often be less severe
in vaccinated children). If 100 children are vaccinated with a full schedule of
pertussis-containing vaccines, 20 of the children might still get whooping cough
but, once again, the disease is often less severe in these vaccinated children. To
put it another way, if you do not vaccinate 100 children with MMR vaccine, and
the children are exposed to measles, all of them will catch the disease with a

358 The Australian Immunisation Handbook 9th Edition

risk of high rates of complications like pneumonia or encephalitis. The reason


why fewer children become infected than these figures suggest is due to the
high vaccine coverage rates in the community. If there are high coverage rates,
there is less chance of contact with the infection and, although children may be
susceptible, they have a low chance of contact with the infection.

(iv) What about homeopathic immunisation?


Homeopathic immunisation has not been proved to give protection against
infectious diseases; only conventional vaccination produces a measurable
immune response. The Council of the Faculty of Homeopathy, London, issued a
statement in 1993, which reads: The Faculty of Homeopathy, London, strongly
supports the conventional vaccination program and has stated that vaccination
should be carried out in the normal way, using the conventional tested and
proved vaccines, in the absence of medical contraindications.51

4. Where can I get more information about vaccination?


More information about vaccination can be found in the following publications
produced by the Australian Government Department of Health and Ageing:
Understanding childhood immunisation
Immunisation myths and realities responding to arguments against immunisation:
a guide for providers
The following two websites include further publications, fact sheets, etc. and are
recommended for both immunisation service providers and the general public:
Immunise Australia website http://www.immunise.health.gov.au

Also, check with your local State or Territory public health unit or local council,
maternal child health nurse, or public health vaccination clinic for more
information (see Appendix 1, Contact details for Australian, State and Territory
Government health authorities and communicable disease control).

References
Full reference list available on the electronic Handbook or website
http://immunise.health.gov.au.

Appendix 5 359

Appendices

The National Centre for Immunisation Research and Surveillance of Vaccine


Preventable Diseases (NCIRS) website www.ncirs.usyd.edu.au

Appendix 6: Definitions of adverse


events following immunisation
Notify any events that the reporter considers serious and which may be related
to the vaccine or vaccines administered. See Section 1.5.2, Adverse events
following immunisation.
Abscess
Occurrence of a fluctuant or draining fluid-filled lesion at the site of injection,
with or without fever.
Bacterial: purulent collection.
Sterile abscess: no evidence of bacterial infection.
Acute flaccid paralysis [diagnosis must be made by a physician]
Acute onset of flaccid paralysis of one or more limbs following any vaccine.
Allergic reaction (generalised)
A non-anaphylactic, generalised reaction characterised by 1 or more symptoms
or signs of skin and/or gastrointestinal tract involvement WITHOUT respiratory
or cardiovascular involvement.
(NB. See also Anaphylaxis).
Anaphylaxis
A rapidly evolving generalised multi-system allergic reaction characterised by
1 or more symptoms or signs of respiratory and/or cardiovascular involvement
AND involvement of other systems such as the skin or gastrointestinal tract.
Respiratory: difficulty/noisy breathing, swelling of the tongue, swelling/
tightness in the throat, difficulty talking/hoarse voice, wheeze or
persistent cough.
Cardiac: loss of consciousness, collapse, pale and floppy (babies),
hypotension.
Arthralgia
Joint pain without redness or swelling.
Arthritis
Joint pain with redness and/or swelling.
Brachial neuritis
Pain in arm causing persisting weakness of limb on side of vaccination.
Death
Any death of a vaccine recipient temporally linked to vaccination, where no
other clear cause of death can be established.

360 The Australian Immunisation Handbook 9th Edition

Disseminated BCG
Disseminated infection occurring after BCG vaccination and confirmed by
isolation of Mycobacterium bovis BCG strain.
Encephalopathy [diagnosis must be made by a physician]
Encephalopathy is an acute onset of major neurological illness temporally
linked with vaccination and characterised by any 2 or more of the following 3
conditions:
seizures,
severe alteration in level of consciousness or mental status (behaviour and/or
personality) lasting for 1 day or more, and/or
focal neurological signs which persist for 1 day or more.
Encephalitis [diagnosis must be made by a physician]
Encephalitis is characterised by the above-mentioned symptoms and signs
of cerebral inflammation and, in many cases, CSF pleocytosis and/or virus
isolation.
Extensive limb swelling
Swelling of the limb, with or without redness, which:
extends from the joint above to the joint below the injection site, or beyond a
joint (above or below the injection site), or
results in the circumference of the limb being twice the normal size.
Faint
See Vasovagal episode.

Guillain-Barr Syndrome (GBS) [diagnosis must be made by a physician]


Acute onset of rapidly progressive, ascending, symmetrical flaccid paralysis,
without fever at onset of paralysis and with or without sensory loss. Cases
are diagnosed by cerebrospinal fluid (CSF) investigation showing dissociation
between cellular count and protein content.
Hypotonichyporesponsive episode (shock, collapse)
The sudden onset of pallor or cyanosis, limpness (muscle hypotonia), and
reduced responsiveness or unresponsiveness occurring after vaccination,
where no other cause is evident such as a vasovagal episode or anaphylaxis.
The episode usually occurs 1 to 48 hours after vaccination and resolves
spontaneously.

Appendix 6 361

Appendices

Fever
Only very high fever should be reported, eg. >40.5C.

Injection site reaction (severe)


Reaction (redness and/or swelling) at site of injection which:
persists for more than 3days AND is associated with ongoing symptoms
such as pain or an inability to use the limb (see Brachial neuritis above), and
does not fulfil the case definition for extensive limb swelling (see Extensive
limb swelling above), or
requires hospitalisation.
Intussusception [diagnosis must be made by a hospital physician]
The invagination of a proximal segment of bowel into the distal bowel lumen.
Lymphadenitis (includes suppurative lymphadenitis)
Occurrence of either:
at least 1 lymph node, 1.5 cm in diameter or larger, or
a draining sinus over a lymph node.
Meningitis [diagnosis must be made by a physician]
Acute onset of major illness with fever and often neck stiffness/positive
meningeal signs (Kernig, Brudzinski) and with CSF pleocytosis.
Nodule
A discrete or well demarcated soft tissue mass or lump that is firm and is at the
injection site in the absence of abscess formation, warmth and erythema.
Orchitis
Swelling with pain and/or tenderness of testes.
Osteitis
Inflammation of the bone due to BCG vaccination.
Osteomyelitis
Proven bacterial infection of bone.
Parotitis
Swelling and/or tenderness of parotid gland or glands.
Rash
Severe or unusual rash.
Screaming (persistent)
The presence of crying which is continuous and unaltered for longer than 3hours.
Seizure
Witnessed sudden loss of consciousness and generalised, tonic, clonic, tonicclonic, or atonic motor manifestations.
febrile seizures: with fever 38.5C,
afebrile seizures: without fever,
syncopal seizures: syncope/vasovagal episode followed by seizure(s).
362 The Australian Immunisation Handbook 9th Edition

Sepsis
Acute onset of severe, generalised illness due to bacterial infection and confirmed
by positive blood culture.
Subacute sclerosing panencephalitis [diagnosis must be made by a physician]
Degenerative central nervous system (CNS) condition with laboratory
confirmation of abnormal serum and CSF measles antibodies.
Syncope
See Vasovagal episode.
Thrombocytopenia
Platelet count <50 x 109/L.
Toxic shock syndrome [diagnosis must be made by a physician]
Abrupt onset of fever, vomiting, watery diarrhoea and shock within a few hours
of vaccination as can be associated with other conditions listed here.
Vaccine-associated paralytic poliomyelitis
See Acute flaccid paralysis.
Vasovagal episode (syncope, faint)
Episode of pallor and unresponsiveness or reduced responsiveness or feeling
light headed AND
occurring while vaccine being administered or shortly after (usually within
5minutes), AND
bradycardia, AND

(See Table 1.5.1 to distinguish from anaphylaxis).


Other severe or unusual events
Any unusual event that does not fit into any of the categories listed above, but
is of medical or epidemiological interest, should be reported with a detailed
description of the clinical features.
Report by telephone to State or Territory Health Department or notify by the blue
card to ADRAC (see Section 1.5.2, Adverse events following immunisation).
Note: The Brighton Collaboration is an international group considering
definitions of adverse events following immunisation. Its website is:
http://www.brightoncollaboration.org.

Appendix 6 363

Appendices

resolution of symptoms with change in position (supine position or head


between knees or limbs elevated).

Appendix 7: Glossary of technical terms


Adjuvant
a preparation which may be added to a vaccine to improve the immune response
to that vaccine.
ADT
adult diphtheria and tetanus vaccine (also referred to as dT). Trade name used
for diphtheria-tetanus vaccine previously made by CSL for use in adults.
Adverse event following immunisation (AEFI)
an unwanted reaction following administration of a vaccine, which may or may
not be caused by the vaccine; adverse events may be at the site of injection, or
may be a general illness or a general allergic reaction.
Anaphylaxis
a sudden and severe allergic reaction, which results in a serious fall in blood
pressure and/or respiratory obstruction and may cause unconsciousness and
death if not treated immediately.
Attenuation
the process of modifying a virus or bacteria to reduce its virulence (diseaseinducing ability) while retaining its ability to induce a strong immune response
(immunogenicity).
Bacteria
microorganisms that are smaller than a blood cell but bigger than a virus; examples
of bacterial infections are diphtheria, tetanus, pertussis, Hib and tuberculosis.
BCG
Bacillus of Calmette-Gurin, a vaccine that protects against tuberculosis.
Carrier
a person who has an infection which, although not necessarily causing symptoms,
may still be active and may spread to others; the carrier state may last foryears;
examples of infections that can result in the carrier state are hepatitis B and typhoid.
Conjugate
some bacterial vaccines (eg. Hib and pneumococcal conjugate vaccines) are made
from the chemical linking (conjugation) of a tiny amount of the sugar (correctly
known as the polysaccharide) that makes up the cell coat of the bacteria with a
protein molecule, in order to improve the immune response to the vaccine.
Contraindication
a reason why a vaccine or drug must not be given.
Corticosteroid
a drug used to reduce inflammation and other immune responses.

364 The Australian Immunisation Handbook 9th Edition

dT
diphtheria-tetanus vaccine for use in adults (ADT).
DTP/DTPa
a vaccine that protects against diphtheria, tetanus and pertussis (whooping
cough). The DTP used in Australia and many other industrialised countries
is DTPa, which contains an acellular pertussis component made of refined
pertussis extracts instead of inactivated whole pertussis bacteria. The acronym
DTPa, using capital letters, signifies child formulations of diphtheria, tetanus
and acellular pertussis-containing vaccines, and denotes the substantially larger
amounts of diphtheria toxoid and pertussis antigens in these formulations than
in the adolescent/adult formulations.
dTpa
adolescent/adult formulation diphtheria-tetanus-acellular pertussis vaccine.
dTpa contains substantially lower concentrations of diphtheria toxoid and
pertussis antigens than the child formulations (which are signified by using all
capital letters (DTPa)).
Effectiveness
the extent to which a vaccine produces a benefit in a defined population in
uncontrolled or routine circumstances.
Efficacy
the extent to which a vaccine produces a benefit in a defined population in
controlled or ideal circumstances.

Encephalopathy
a general term to describe a variety of illnesses that affect the brain, including
encephalitis.
Endemic
endemic infections are present all the time in a community.
Epidemic
epidemic infections are those that spread rapidly in a community; measles and
influenza viruses are common causes of epidemics in Australia; small epidemics
are often called outbreaks.
Febrile
related to a fever, as in febrile illness and febrile convulsions.
HAV
abbreviation for hepatitis A virus, the cause of hepatitis A, a common food-borne
infection in travellers to developing countries.

Appendix 7 365

Appendices

Encephalitis
inflammation of the brain.

HBsAg
hepatitis B surface antigen; a marker in the blood that indicates that the person is
a carrier of active hepatitis B virus infection.
HBV
abbreviation for hepatitis B virus, a virus that is spread in body fluids in various
ways including blood-to-blood contact through sharing injection equipment, and
through sexual intercourse.
Hepatitis
an inflammation of the liver; can be caused by viral infections.
Hib
Haemophilus influenzae type b; a bacterium that causes meningitis and other
serious infections in young children.
HIV
human immunodeficiency virus, or the AIDS virus; people with HIV infection
may have weakened immunity and need special vaccinations to protect them
against other infections.
Human papillomavirus
a group of viruses, some of which have been associated with some forms of
cervical cancer; some can also cause genital warts.
Hypotonic-hyporesponsive episode (HHE)
a rare adverse event which may follow some hours after DTPa vaccination; the
child becomes pale, limp and unresponsive; the condition may last from a few
minutes to hours but causes no long-term serious problems.
Immunisation
the process of inducing immunity to an infectious agent by administering a
vaccine.
Immunity
the ability of the body to fight off certain infections; immunity can result from
natural (wild) infections or from vaccination.
Immunogenicity
the ability (or the degree) to which a particular substance, in this context a
vaccine, may provoke an immune response.
Immunoglobulin
a protein extract from blood, sometimes called antibody, which fights off
infection; injection of immunoglobulins provides temporary immunity against
certain infections.

366 The Australian Immunisation Handbook 9th Edition

Incubation period
after a person is infected with bacteria or viruses, it often takesdays orweeks
for the infection to cause an obvious illness; the time between exposure to the
infectious agent and development of the disease is called the incubation period.
Infection
an infection occurs when bacteria or viruses invade the body; if the body cannot
fight the infection, it may cause an illness.
Intradermal injection
an injection into the surface layers of the skin; this is used for the administration
of BCG, the tuberculosis vaccine.
Intramuscular (IM) injection
an injection into the muscle; vaccines are usually injected into a muscle of the
upper outer thigh, or a muscle in the upper arm.
IPV
inactivated poliomyelitis vaccine; an injectable vaccine formerly known as Salk
vaccine.
Invasive disease
this term is often used when talking about pneumococcal or meningococcal
disease. This term means that the bacteria (or germs) have been found in the
blood, spinal fluid or another part of the body which would normally be sterile
(or germ free).
Appendices

Jaundice
yellow skin colour that may result from severe hepatitis.
JE
Japanese encephalitis; a viral encephalitis.
MMR
measles-mumps-rubella vaccine.
MMRV
measles-mumps-rubella-varicella vaccine.
OPV
oral poliomyelitis vaccine; also known as Sabin vaccine. This vaccine is no longer
routinely used in Australia.
Pandemic influenza
a global epidemic that results when a new strain of influenza virus appears in the
human population. It causes more severe disease in the population because there
is little immunity to this new strain.

Appendix 7 367

Paracetamol
a medicine that helps reduce fever; it may be given to minimise fevers following
vaccination.
Pertussis
whooping cough, an illness caused by a bacterium, Bordetella pertussis.
Polysaccharide
a group of complex carbohydrates (sugars) which make up the cell coating
present in some bacteria.
Polyvalent vaccine
a combination vaccine which protects against more than one disease; examples
are DTPa and MMR.
PRP-OMP
a type of Hib vaccine.
PRP-T
a type of Hib vaccine.
Rotavirus
a virus that is a common cause of diarrhoea (and often vomiting as well) in
young children. The diarrhoea can be severe in very young children, such that
they may need intravenous fluids (ie. through a vein in the arm) in hospital.
Rubella
a viral illness, sometimes also known as German measles.
Subcutaneous (SC) injection
an injection into the tissue between the skin and the underlying muscle.
Vaccination
the administration of a vaccine; if vaccination is successful, it results in immunity.
Vaccine
a product often made from extracts of killed viruses or bacteria, or from live
weakened strains of viruses or bacteria; the vaccine is capable of stimulating an
immune response that protects against natural (wild) infection.
Varicella
chickenpox, an infection caused by the varicella-zoster virus.
Virus
a tiny living organism, smaller than a bacterium, that can cause infections;
measles, rubella, mumps, polio, influenza and hepatitis B are examples of viruses.
Zoster
an abbreviation for herpes zoster infection (also known as shingles); a painful
rash and illness caused by the varicella-zoster (chickenpox) virus.

368 The Australian Immunisation Handbook 9th Edition

Appendix 8: List of commonly


used abbreviations
ABL

Australian bat lyssavirus

ACIP

Advisory Committee on Immunization Practices

ACIR

Australian Childhood Immunisation Register

ADRAC

Adverse Drug Reactions Advisory Committee

AEFI

adverse event following immunisation

AFP

acute flaccid paralysis

AIDS

acquired immunodeficiency syndrome

anti-HBs

hepatitis B surface antibody

ATAGI

Australian Technical Advisory Group on Immunisation

BCG

Bacillus of Calmette-Gurin

BSE

bovine spongiform encephalopathy

CCM

cold chain monitor

CDT

diphtheria-tetanus vaccine for children (no longer in use)

cm

centimetre

CSF

cerebrospinal fluid

dT

diphtheria-tetanus vaccine for use in adults

DTPa child formulation diphtheria-tetanus-acellular pertussis vaccine

ELISA/EIA

enzyme-linked immunosorbent assay

GVHD

graft versus host disease

HAV

hepatitis A virus

HBIG

hepatitis B immunoglobulin

HBsAg

hepatitis B surface antigen

HBV

hepatitis B virus

HCW

Healthcare worker

HepA

hepatitis A

HepB

hepatitis B

HHE

hypotonic-hyporesponsive episode

Hib

Haemophilus influenzae type b

HIV

human immunodeficiency virus

HPV

human papillomavirus

Appendix 8 369

Appendices

dTpa adolescent/adult formulation diptheria-tetanus-acellular


pertussis vaccine

HRIG

human rabies immunoglobulin

HSCT

haematopoietic stem cell transplant

HZ

herpes zoster

IM

intramuscular

IMD

invasive meningococcal disease

IPD

invasive pneumococcal disease

IPV

inactivated poliomyelitis vaccine

ITP

idiopathic thrombocytopenia purpura

IU

international units

IV

intravenous

JE

Japanese encephalitis

kg

kilogram

ng

nanogram

microgram

mg

milligram

mL

millilitre

mm

millimetre

4vMenPV

meningococcal polysaccharide vaccine (tetravalent)

MenCCV

meningococcal C conjugate vaccine

MMR

measles-mumps-rubella

MMRV

measles-mumps-rubella-varicella

MS

multiple sclerosis

NHIG

normal human immunoglobulin

NHMRC

National Health and Medical Research Council

NIP

National Immunisation Program

OMP

outer membrane protein

OPV

oral poliomyelitis vaccine (no longer in use)

PHN

post-herpetic neuralgia

PI

product information

7vPCV

7-valent pneumococcal conjugate vaccine

PPD

purified protein derivative

23vPPV

23-valent pneumococcal polysaccharide vaccine

PRP

polyribosylribitol phosphate

SC

subcutaneous

370 The Australian Immunisation Handbook 9th Edition

SIDS

sudden infant death syndrome

SOT

solid organ transplant

TB

tuberculosis

TGA

Therapeutic Goods Administration

TIG

tetanus immunoglobulin

VAPP

vaccine-associated paralytic poliomyelitis

vCJD

variant Creutzfeldt-Jakob disease

VV

varicella vaccine

VZV

varicella-zoster virus

WHO

World Health Organization

ZIG

zoster immunoglobulin

Appendices

Appendix 8 371

Appendix 9: Dates when vaccines


became available in Australia
Public sector
Australia

Vaccine

Exceptions

1945-46

Tetanus toxoid

1953

DTPw (diphtheria/tetanus/pertussis whole cell)

1956 May

Poliomyelitis (SALK)

1966 Sep

Poliomyelitis (OPV) (oral Sabin)

1969

Measles

1971 Feb

Rubella (adolescent girls)

1975

CDT (child diphtheria/tetanus)

1981 Jul

Mumps

1982

ADT (adult diphtheria/tetanus)

1983

Measles/Mumps

1986

CDT-DTP 4th dose introduced (1st pertussis booster)

1987 Nov

NT 1988 Jan
SA 1996

Hepatitis B (for at-risk infants)

1989

SA 1996

MMR (infant dose)

1992 May

Hib (for children 18months to 5years of age)

1993 Apr

Hib (all infants born from Feb 1993)

1993 Jul

Hepatitis A (Havrix) (unfunded)

1994

MMR (males and females in Grade 6)

1995

CDT-DTP 5th dose introduced (2nd pertussis booster)

1997 Oct

Tas 1997 Oct


Qld 1997 Dec

Influenza (program for over 65s)

1997
1998 Jan

DTP acellular (Infanrix) boosters for infants aged


18months and 45years to replace DTP 4th and 5th doses

Qld 1998 Mar


Tas 1998 Mar
NT 1998 Apr
NSW 1999
SA 1999

Hepatitis B (adolescent dose)

1998

MMR (Primary School program)

1998

MMR/OPV 4-year-old booster (DTP, MMR,OPV)


program before commencing school

372 The Australian Immunisation Handbook 9th Edition

Public sector
Australia

Pneumococcal polysaccharide 23-valent


vaccine (over 65s) (unfunded)

1998
1999 Feb

Vaccine

Exceptions

NT 1997 Aug
SA 1997 Aug
Qld 1999 Apr

DTPa (Infanrix) for infants aged 2, 4 and 6months

1999

MMR (1830-year-old program)

2000

COMVAX (Hep B/Hib) available

2000

Adult diphtheria/tetanus 10-yearly boosters ceased

2000 May

NT 1990 Aug

Hepatitis B (universal infant dose)


Hepatitis B booster doses no longer recommended

2001

Pneumococcal conjugate 7-valent vaccine (for


Aboriginal and Torres Strait Islander children
and all children in Central Australia only)

2001 Dec

Meningococcal C conjugate vaccine (Meningitec) (unfunded)

2001

Varicella (chickenpox) (unfunded)

2001

HibTITER vaccine ceased

2002 Aug

Meningococcal C conjugate vaccine (NeisVac-C) (unfunded)

2002 Oct

Meningococcal C conjugate vaccine (Menjugate) (unfunded)

2003 Sep

DTPa 4th dose at 18months of age ceased

2003 Jan

Meningococcal C conjugate vaccine (at 12months of


age and catch-up until May 2007 for 119-year-olds)

2003 Sep

Pneumococcal conjugate 7-valent vaccine (for


children with medical risks <5years of age)

2004 Jan

dTpa (Boostrix) for 1517years (Year 10


school program) replaced ADT

2004 Sep

Combination vaccines with 4, 5 and 6 antigens available

2005 Jan

Pneumococcal conjugate 7-valent vaccine (for


infants aged 2, 4 and 6months and medical at-risk
children plus catch-up in 2005 for children born
between 1 January 2003 and 31 December 2004)

2005 Jan

Pneumococcal polysaccharide 23-valent


vaccine (funded for adults aged >65years)

2005 Nov

Inactivated polio (IPV) vaccine (given in


combination with DTPa scheduled at 2, 4 and
6months and 4years in place of OPV)

Appendix 9 373

Appendices

2000

Public sector
Australia

Vaccine

Exceptions

2005 Nov

Hepatitis A vaccine (for all Indigenous children


aged 5years living in Queensland, the Northern
Territory, Western Australia and South Australia)

2005 Nov

Varicella vaccine scheduled at 18months and 13years of age

2007 Apr

Human papillomavirus vaccine (for girls aged


1213years plus a 2-year catch-up period to end
of June 2009 for girls aged 1426years)

2007 May

NT 2006 Oct

Rotavirus vaccine (for all children born from 1 May 2007)

374 The Australian Immunisation Handbook 9th Edition

Appendix 10: Summary table procedures


for a vaccination encounter
This table summarises the information provided in Chapters 1.31.5 and provides
an overview of the requirements before, during and after a vaccination encounter.
This table can also be photocopied and used as an audit tool, if required.
Pre-vaccination procedures (Chapter 1.3)

Prepare anaphylaxis response kit: check availability of the protocols,


equipment and drugs necessary for the management of anaphylaxis, before
each vaccination session. (1.3.1)
Only vaccine that has been transported and stored at the correct cold chain
temperature of between +2C to +8C should be administered. Follow the
National Vaccine Storage Guidelines: Strive for 5. (1.3.2)
Perform pre-vaccination screening to determine the persons medical
fitness for vaccination, and possible need for additional vaccines. Any
concern about the persons eligibility for vaccination must be discussed
with a medical practitioner, paediatrician or public health physician with
expertise in vaccination (see Appendix 1 for phone numbers for State/
Territory health authorities.) If a persons health status or suitability for
vaccination cannot be determined, defer vaccination and seek advice. (1.3.4)

Obtain valid consent from the person to be vaccinated, or that persons


parent/carer: this includes providing the appropriate information about
the risks and benefits of vaccination and the risks of vaccine-preventable
diseases. (Written vaccination information can be provided to parents as
early as the last trimester of pregnancy or at the well-baby check.) Advise
the person to be vaccinated, or the parent/carer of a child, of the incidence of
common adverse events that may occur following vaccination. This advice
and the parents consent should be documented. It is important that the
parent be given a contact phone number in case a significant adverse event
occurs within 24 to 48 hours of the vaccination. (1.3.3)

Appendix 10 375

Appendices

Review the individuals vaccination history and, based on documented


evidence, decide on the appropriate vaccine(s) to be administered. If the
recommended vaccination schedule for age has not been completed, plan
and document a catch-up schedule and discuss this with the person or
parent/carer. (1.3.5)

Administration of vaccines (Chapter 1.4)

Follow standard occupational health and safety guidelines to minimise


the risk of needle-stick injury. (1.4.1)
Depending on the vaccine(s) that are to be administered, and the age and
size of the person to be vaccinated, decide on the appropriate injection site
and route, and the injection equipment required (ie. syringe size, needle
length and gauge) as recommended in the current NHMRC immunisation
guidelines. Use a new, sterile, disposable syringe and needle for each
injection. (1.4.21.4.6)
Prepare the vaccine (check whether the vaccine is injectable or oral):
Check each individual dose (ie. ampoule, pre-filled syringe or vial) to see
that the expiry date has not lapsed, and that there is no particulate
matter or colour change in the vaccine.
Reconstitute the vaccine as needed immediately before administration,
preferably using a separate needle to draw up the diluent or as
recommended by the manufacturer. Use only the diluent supplied with
the vaccine. Mix fully, and draw up the vaccine.
Locate the injection site by fully uncovering the appropriate limb(s) and
visualising the correct anatomical markers. Position the limb for vaccination
so that the muscles are relaxed (usually a flexed position). Keep the limb as
immobile as possible without using excessive restraint. Ensure that the skin
is visibly clean. (1.4.71.4.8)
Administer the vaccine(s) using the recommended technique (IM,
SC or Oral). For injectable vaccines, follow the recommendations for
administering more than 1 vaccine into a limb during the encounter. Do not
inject oral vaccines. Remove the needle briskly after IM injection. (1.4.5, 1.4.9)

376 The Australian Immunisation Handbook 9th Edition

Post-vaccination procedures (Chapter 1.5)

Immediate after-care
Dispose of used needles, syringes and vaccine vials/ampoules in
accordance with standard infection control guidelines.
Cover the puncture wound quickly with a dry cotton wool ball and
hypoallergenic tape as needed. Apply gentle pressure for 12 minutes
but do not massage.
Remove the cotton wool and tape after a few minutes.
Continue using comfort and distraction techniques to alleviate any
distress andpain. Note:paracetamol is not used routinely at the time
of vaccination but may be recommended as required for fever or pain.
(1.5.1)
Managing adverse reactions, documentation and follow-up
Remind the vaccinated person, or the parent/carer of a child, about the
possible common adverse events following immunisation and how to
manage them. It is preferable to provide this as written information (see
inside back cover of this Handbook).
Before departure, inform the person or the parent/carer, preferably in
writing, of the date of the next scheduled vaccination.

Take the opportunity to check the vaccination status of other


family members (as appropriate) and provide (or refer) for catch-up
vaccination.
Document the details of vaccination:

(i) on a record to be retained by the person, or the parent/carer of a child,

(ii) on the relevant clinical record (electronic or hard-copy), and

(iii) on an ACIR (or equivalent) encounter form, for children <7years of age.

Remind the vaccinated person, or the parent/carer of a child,


to promptly report any significant adverse event following
immunisation to the vaccinator, so that it can be reported to either the
Adverse Drug Reactions Advisory Committee (TAS) or to the relevant
State/Territory health authorities (ACT, NSW, NT, QLD, SA, VIC and
WA). (1.5.21.5.4)

Appendix 10 377

Appendices

The vaccinated person and/or parent/carer should be advised to


remain in a nearby area for a minimum of 15 minutes after the
vaccination. The area should be close enough to the vaccinator, so that
the child/person can be observed and medical treatment can be readily
obtained if needed.

Index
A

abattoir workers, 107, 258, 260


abbreviations list, 36971
ABL see Australian bat lyssavirus (ABL)
infection
Aboriginal and Torres Strait Islander
people, 704
children and infants,
BCG vaccine, 701, 300
catch-up, 32, 345
Haemophilus influenzae type b, 71,
132, 135
hepatitis A, 72, 144
pneumococcal disease, 345, 72,
2401, 243, 247
hepatitis A, 142
influenza, 190
Japanese encephalitis (JE), 73
pneumococcal disease, 73, 2401, 246
service delivery to, 734
tuberculosis, 297
ACIR, 67 seealso Australian Childhood
Immunisation Register
Adacel, 42, 126, 230, 290 seealso dTpa
Adacel Polio, 42, 126, 230, 254, 290
seealso dTpa-IPV
additives in vaccines, 341, 3567
adjuvants, 341, 356
administration see dosage and
administration
adoption of children overseas, 159
adrenaline, 64
ADT Booster, 42, 126, 290 seealso dT;
diphtheria; tetanus
Adverse Drug Reactions Advisory
Committee (ADRAC), 65
adverse events following immunisation
(AEFI), 5866
cholera vaccine, 123
definition(s) of, 3603, 364
diphtheria-containing vaccine, 128
Haemophilus influenzae type b
vaccine, 137
hepatitis A vaccine, 146

hepatitis B vaccine, 1623


how to report, 645
human papillomavirus (HPV)
vaccine, 1734
immunoglobulins, 17781
in children and infants, 84
influenza vaccine, 1934
Japanese encephalitis (JE) vaccine,
199200
management of, 614
measles-containing vaccine, 2089
meningococcal vaccine, 220
mumps-containing vaccine, 225
pertussis-containing vaccine, 2345
pneumococcal vaccine, 247, 249
poliomyelitis vaccine, 255
Q fever vaccine, 2623
rabies vaccine, 118
reporting, 645
rotavirus vaccine, 269, 2723
rubella vaccine, 2801
smallpox vaccine, 2856
tetanus-containing vaccine, 295
tuberculosis vaccine, 301
typhoid vaccine, 307
varicella vaccine, 3178
yellow fever vaccine, 327
Afghanistan, 77
Africa
avian influenza, 184
cholera, 120
hepatitis B, 77, 150
measles, 202
meningococcal disease, 79, 214
poliomyelitis, 251
rabies, 110, 115
rubella, 278
yellow fever, 75, 79, 322, 324, 326
aged see older people
agricultural college staff and students,
107, 258, 260
allergy, 205, 279, 316, 3501 seealso egg
protein allergy
aluminium in vaccines, 341, 356
ambulance personnel, 105, 159, 192
Americas
cholera, 120
poliomyelitis, 251

378 The Australian Immunisation Handbook 9th Edition

rabies, 110, 115, 119


rubella, 278
tuberculosis, 297
typhoid, 304
aspirin therapy, 191, 207, 318
asplenia (functional or anatomical), 16,
19, 36, 87, 93, 1001, 136, 138, 213, 218,
244, 2467
asthma, 61, 191, 350, 353
Australian bat lyssavirus (ABL)
infection, 107, 1109 seealso rabies
post-exposure prophylaxis,
vaccine, 42, 1146
rabies immunoglobulin (HRIG),
1167
pre-exposure prophylaxis, 1124
Australian Childhood Immunisation
Register (ACIR), 223, 679, 202, 332
Australian Red Cross Blood Service
(ARCBS), 176, 319
Australian Standard Vaccination
Schedule (ASVS), 3
Australian Technical Advisory Group on
Immunisation (ATAGI), 334
autism, 61, 209, 352
autistic spectrum disorder, 209, 352
autoimmune diseases, 1012 seealso
impaired immunity, individuals with
Avaxim, 42, 140 seealso hepatitis A (HAV)
avian influenza, 184, 1867
azithromycin, 2356, 238, 239

babies (neonates) seealso preterm babies


bronchopulmonary dysplasia (BPD),
182
hepatitis B, 1567, 162, 163
influenza, 185
measles, 211
pertussis, 236, 237, 238
respiratory syncytial virus (RSV), 182
tetanus, 288, 289
tuberculosis, 300
varicella, 309
bacteraemia, 240
Bali
Japanese encephalitis (JE) virus in,
198

Index 379

Index

rabies, 110, 115


rubella, 278
yellow fever, 75, 322, 323, 324
anaesthesia, 104
anaphylactoid see anaphylaxis;
contraindications
anaphylaxis, 614 seealso
contraindications
adrenaline use, 64
differentiation from vasovagal
episode, 62
management of, 634
pre-vaccination screening, 17
signs of, 62
anaphylaxis response kit, 8, 375
animals
human rabies from, 1101
occupational risks and, 107
Q fever from, 258, 260
anthrax, 3
antibiotics
diphtheria, 128
false contraindications and, 21, 348
haematopoietic stem cell
transplantation and, 97
Haemophilus influenzae type b, 1378
manufacturing residuals, 341, 357
meningococcal disease, 220
pertussis, 2359
smallpox, 285
splenic dysfunction and, 101
tetanus, 294
typhoid vaccine, oral and, 83, 305,
307
antibody deficiency disorders, 180
anti-HAV IgG, 139, 142
anti-HAV IgM, 139, 142, 146
anti-HBs, 31, 90, 149, 153, 157, 158, 1601
armed forces personnel, 106, 159, 297,
306 seealso travellers
Asia
avian influenza, 184
cholera, 120
hepatitis B, 77, 150
Japanese encephalitis (JE), 75, 78, 82,
195, 1978, 199
measles, 358
meningococcal disease, 214

rabies in, 119


bats see Australian bat lyssavirus (ABL)
infection
BCG vaccine, 298 seealso tuberculosis
bleeding disorders, 104, 159, 178, 2078
blood and organ donors, 150, 159, 175
blood products/transfusions, 1023, 159,
206, 271, 280, 317
live vaccines and, interval between,
1023
body-piercers, 107, 159
bone marrow transplantation
see haematopoietic stem cell
transplantation (HSCT)
boosters and revaccination
diphtheria, 127, 128, 129
Haemophilus influenzae type b, 134,
136, 138
hepatitis A, 142, 145
hepatitis B, 156, 1613
human papillomavirus (HPV), 170
Japanese encephalitis (JE), 198, 200
meningococcal disease, 218
pertussis, 228, 2313, 238, 3467
pneumococcal disease, 243, 246, 247
poliomyelitis, 255
Q fever, 262
rabies, 113
tetanus, 2924
typhoid, 145, 306
Boostrix, 42, 126, 230, 291 seealso dTpa
Boostrix-IPV, 42, 126, 230, 254, 291 seealso
dTpa-IPV
Bordetella pertussis see pertussis
botulism antitoxin, 181
Botulism Immune Globulin (BIG), 181
bovine spongiform encephalopathy
(BSE), 354 seealso mad cow disease
brachial neuritis, 61 seealso adverse
events following immunisation (AEFI)
breakthrough varicella, 3101, 3123
breastfeeding and lactation, 21, 89, 174
seealso pregnancy and vaccination
cholera vaccine and, 123
human papillomavirus (HPV)
vaccine and, 174
meningococcal vaccine and, 222
Q fever vaccine and, 263

rotavirus vaccine, 268


rubella vaccine and, 280
smallpox vaccine and, 285
bronchopulmonary dysplasia (BPD), 182
BSE see bovine spongiform
encephalopathy

cancer chemotherapy, 924


cardiac disease, 190, 246, 247
cardiovascular disease, 122
carers, 106
carriers
hepatitis B, 14950, 151, 156, 157, 158
meningococcal disease, 213, 2201
typhoid, 308
catch-up, 2138
children aged <8,
guidelines, 312
Haemophilus influenzae type b, 33
minimum age, 30
minimum interval, 29
number of doses required, 28
pneumococcal vaccination, 346
worksheet, 27
children aged 8, adolescents and
adults, 378
immigrants, 108
ceftriaxone, 137, 221
CERVARIX, 42, 169 seealso human
papillomavirus (HPV)
cervical cancer, 164, 1678
chemoprophylaxis see antibiotics
chemotherapy, 924
chickenpox see varicella
childcare facilities
Haemophilus influenzae type b, 138
hepatitis A, 147
hepatitis B, 160
measles, 178
pertussis, 232, 236, 237, 238
rubella, 279
childcare workers, 87, 104, 105, 144, 147,
160, 232, 313, 346
children and infants
asplenia, 1001
bleeding disorders, 104
catch-up for, 223, 2530, 338

380 The Australian Immunisation Handbook 9th Edition

hepatitis B-containing, 152


measles, mumps and rubella, 203,
224, 277
pertussis-containing, 22930
poliomyelitis-containing, 2534
tetanus-containing, 28991
typhoid-containing, 304
communicability period see infectious
period
communicable disease control, telephone
contact details for, 333
compatibility see interchangeability of
vaccines
compulsory vaccination, 347
COMVAX, 31, 42, 71, 133 seealso
Haemophilus influenzae type b
concurrent vaccination see simultaneous
administration
congenital rubella syndrome (CRS),
2745, 278
congenital varicella syndrome, 309
conjugate vaccines see Haemophilus
influenzae type b; meningococcal
disease; pneumococcal disease
consent, 124, 375
contacts (disease)
diphtheria, 1289
Haemophilus influenzae type b, 1378
hepatitis A, 1467
hepatitis B, 158
measles, 20910, 211
meningococcal disease, 213, 218, 219,
2201
pertussis, 232, 2378
rotavirus, 271
varicella, 314, 318
contraindications, 14, 20, 60, 34751
anaphylaxis and, 20, 3478
BCG vaccine, 300
cancer patients, 924
cholera vaccine, 123
corticosteroids, 92, 118, 205, 207,
3489
diphtheria-containing vaccine, 128
false, 21
Haemophilus influenzae type b
vaccine, 137
hepatitis A vaccine, 146

Index 381

Index

consent, 123
deceased, notification to ACIR of, 68
HIV-infected, 99
injection sites, 456, 345
living overseas, 68
multiple injections, 24, 567, 345
positioning for vaccination, 4751
vaccination after AEFI, 84
with intercurrent illnesses, 348
cholera, 42, 78, 1203
adverse events, 123
contraindications, 123
dosage and administration, 122
precautions, 123
pregnancy, 87, 123
recommendations, 122
chronic conditions and vaccination, 350
chronic graft-versus-host disease
(cGVHD), 97
chronic inflammatory demyelinating
polyneuropathy, 180
hepatitis A, 144
hepatitis B, 150, 158
liver disease, 144, 145, 159
neurological conditions see
neurological disease
renal disease, 90, 191, 246, 247
respiratory conditions, 1901, 246,
247
cidofovir, 2867
ciprofloxacin, 231
clarithromycin, 2356, 239
Clostridium tetani see tetanus
CMV Immunoglobulin-VF (human),
182 seealso cytomegalovirus;
immunoglobulins
coadministration see simultaneous
administration
cold chain, 812
combination vaccines
diphtheria-containing, 1256
dT, 126, 290
DTPa-containing, 1256, 152, 134,
253, 22930, 28991
dTpa-containing, 126, 254, 2901,
Haemophilus influenzae type
b-containing, 134
hepatitis A-containing, 141

hepatitis B vaccine, 162


HIV-infected individuals, 20, 99, 349
human papillomavirus (HPV)
vaccine, 173
immunoglobulins, 178, 180
individuals with impaired immunity,
91, 349
influenza vaccine, 193
Japanese encephalitis (JE) vaccine,
199
live attenuated vaccines and, 201,
91
measles-containing vaccine, 2056
meningococcal vaccine, 219
mumps-containing vaccine, 2056
pertussis-containing vaccine, 233,
348
pneumococcal vaccine, 2489
poliomyelitis vaccine, 255
pregnancy, 85, 86, 206
pre-vaccination screening for, 1720
Q fever vaccine, 262
rabies vaccine, 117
rotavirus vaccine, 270
RSV immunoglobulin, 182
rubella vaccine, 27980
smallpox vaccine, 285
solid organ transplant (SOT)
recipients, 946
tetanus-containing vaccine, 295
typhoid vaccine, 3078
varicella vaccine, 3167
yellow fever vaccine, 3256
correctional facility inmates, 108, 159
correctional facility staff, 106, 159, 192
corticosteroids, 16, 20, 92, 118, 205, 207,
246, 279, 300, 317, 3489
Corynebacterium diphtheriae see diphtheria
coughing illness see pertussis
Creutzfeldt-Jakob disease (vCJD), 354
seealso mad cow disease; bovine
spongiform encephalopathy (BSE)
cystic fibrosis, 190, 246
cytomegalovirus seealso
immunoglobulins
CMV immunoglobulin, 1812

day-care see childcare facilities


deltoid area, 545 seealso injection site
dentists and dental students, 105, 145,
159
Department of Health and Ageing, 334,
359
diabetes, 122, 191, 246, 247, 354
diarrhoea, 60, 62, 63, 78, 122, 123, 270,
273, 307, 308
diphtheria, 12430, 358 seealso DT; dT;
dTpa; DTPa
adverse events, 128
contraindications, 128
diphtheria antitoxin, 129
dosage and administration, 127
public health management, 1289
pregnancy, 87, 129
recommendations, 1278
variations from product information,
12930
diplomats see travellers
distraction techniques, 43
doctors see healthcare workers (HCW)
dosage and administration, 3957, 3757
adrenaline, 64
BCG vaccine, 2989
cholera vaccine, 122
diphtheria-containing vaccine, 127
documentation, 66, 377
Haemophilus influenzae type b, 135,
1378
hepatitis A vaccine, 1423
hepatitis B vaccine, 15362
hepatitis B vaccine accelerated
schedule, 156
human papillomavirus (HPV)
vaccine, 171
immunoglobulins, 146, 163, 176, 178,
179, 182, 183, 2102, 282, 2945, 319
influenza vaccine, 18990
injection sites, 456, 512
Japanese encephalitis (JE) vaccine,
197
measles-containing vaccine, 203,
2102
meningococcal vaccine, 2178
multiple injections, 567

382 The Australian Immunisation Handbook 9th Edition

mumps-containing vaccine, 224


pertussis-containing vaccine, 231
pneumococcal vaccine, 243
poliomyelitis vaccine, 254
Q fever vaccine, 260
rabies immunoglobulin (HRIG), 112,
116
rabies vaccine, 112, 1157
rotavirus vaccine, 268
rubella vaccine, 278
smallpox vaccine, 284
tetanus-containing vaccine, 291
travellers, 76, 801, 823
typhoid vaccine, 3056
varicella vaccine, 312, 320
yellow fever vaccine, 324
drug users see injecting drug users
Dryvax, 283 seealso smallpox
dT, 38, 81, 1267, 2924
DT (CDT vaccine), 127, 291, 339
dTpa, 38, 81, 96, 98, 125, 1267, 229, 290
DTPa-containing vaccines, 96, 98,125,
1267, 229, 290
catch-up, 31
DTPa-hepB-IPV, 125, 152, 229, 253, 290
DTPa-hepB-IPV-Hib, 125, 134, 152, 230,
253, 289
DTPa-IPV, 1256, 229, 253 290
dTpa-IPV, 126, 230, 254, 290
Dukoral, 42, 121, 122 seealso cholera

fainting see vasovagal episodes


farmers, 107, 260
Far North Queensland and Torres Strait
Islands, 106, 195, 196, 198
febrile convulsion, 208, 234, 249
febrile illness, acute, 17, 123, 300, 348
febrile seizure see febrile convulsion
fetus see pregnancy and vaccination
fever, 5960, 85, 199, 2078, 220, 223, 234,
249
flu see influenza
Fluad, 42, 187, 189 seealso influenza
Fluarix, 42, 187, 189, 194 seealso influenza
Fluvax, 42, 187, 189 seealso influenza
Fluvirin, 42, 187, 189, 194 seealso
influenza
food-handlers, 120, 147, 303, 308
formaldehyde, 341, 357
frequently asked questions (FAQ),
34459
funeral industry workers see embalmers

Index 383

Index

egg protein allergy, 117, 193, 194, 2078,


262, 325, 327, 341, 348, 3501
elderly people see older people
embalmers, 107, 159, 300
emergency workers, 106, 159, 192
encephalitis, 75, 201, 225, 2867, 318, 327,
365 seealso Japanese encephalitis (JE)
endemic diseases seealso epidemic
diseases
cholera, 120, 122
hepatitis A, 78, 144, 147
hepatitis B, 156, 159
Japanese encephalitis (JE), 195, 198
measles, 201, 205
poliomyelitis, 252, 255
rabies, 78, 110, 111, 113, 115, 117

typhoid, 78, 3034, 305, 306


yellow fever, 79, 3223, 3245, 326
Engerix-B, 42, 151, 156, 340, 343 seealso
hepatitis B (HBV)
epidemic diseases seealso endemic
diseases
influenza, 1846
Japanese encephalitis (JE), 195
meningococcal disease, 214, 219
pertussis, 2278
rubella, 274
yellow fever, 3234
epilepsy see neurological disease
equipment for vaccination, 3941
erythromycin, 2356, 237, 238
essential service providers, 106, 159, 192
Europe
avian influenza, 184
diphtheria, 128
poliomyelitis, 251
rabies, 110
tuberculosis, 297

GARDASIL, 42, 169 seealso human


papillomavirus (HPV)
gastroenteritis, 2656, 270 seealso
rotavirus
gelatin, 341, 356
General Practice Immunisation Incentive
(GPII) scheme, 69, 332
genital warts, 164, 168, 171
global epidemiology
polio eradication, 252
smallpox eradication, 283
tuberculosis, 297
glossary of technical terms, 3648
graft-versus-host disease (GVHD), 97, 98
Guillain-Barr Syndrome (GBS), 16, 18,
162, 180, 1934, 361

haematological malignancy, 93, 240, 246,


247
haematopoietic stem cell transplantation
(HSCT), 978, 137, 138
haemodialysis patients, 158
haemoglobinopathy, 191, 246
Haemophilus influenzae type b, 1318
Aboriginal and Torres Strait Islander
children, 71, 132, 135
adverse events, 137
asplenia (functional or anatomical),
101, 136
catch-up, 31, 33
contraindications, 137
dosage and administration, 135
haematopoietic stem cell
transplantation, 97, 137
pregnancy, 87, 138
preterm babies, 31, 90, 136
public health management, 1378
recommendations, 1357
variation from product information,
138
Hajj, 79, 219
HAV see hepatitis A (HAV)
Havrix, 42, 140, 146 seealso hepatitis A
(HAV)
Havrix Junior, 42, 140 seealso hepatitis A
(HAV)

hay fever, 350


HBcAg, 149
HBeAg, 149
HbOC, 133, 135 seealso Haemophilus
influenzae type b
HBsAg, 149, 150, 157, 162
HBV see hepatitis B (HBV)
H-B-VAX II, 42, 151 seealso hepatitis B
(HBV)
healthcare workers (HCW), 105, 144, 145,
159, 160, 161, 162, 163, 204, 221, 232,
2378, 255, 279, 281, 300, 313, 3156,
321, 346
hepatitis A (HAV), 72, 78, 105, 106, 107,
13948
adverse events, 146
contraindications, 146
dosage and administration, 1423
immunoglobulin, 146, 177
pregnancy, 88, 148
public health management, 1467
recommendations, 1435
hepatitis B (HBV), 77, 135, 144, 14963,
353
adverse events, 1623
catch-up, 31, 38
contraindications, 162
dosage and administration, 1536
immunoglobulin, 151, 157, 163
non-responders, 1601
post-vaccination serology testing,
160
pregnancy, 88, 163
preterm babies, 31, 90
variation from product information,
163
hepatitis B immunoglobulin (HBIG), 151,
157, 163, 176 seealso hepatitis B (HBV)
Hepatitis B Immunoglobulin-VF, 163
seealso hepatitis B (HBV)
herpes zoster (HZ) see zoster
Hib see Haemophilus influenzae type b
Hiberix, 42, 31, 133, 134 seealso
Haemophilus influenzae type b
Hib (HbOC), 133, 135 seealso Haemophilus
influenzae type b
Hib (PRP-D), 133 seealso Haemophilus
influenzae type b

384 The Australian Immunisation Handbook 9th Edition

Hib (PRP-OMP)-hepB, 133 seealso


Haemophilus influenzae type b
Hib (PRP-T), 133, 134 seealso Haemophilus
influenzae type b
Hirschsprungs disease, 270
HIV-infected individuals, 99100, 122,
146, 158, 191, 199, 205, 206, 34950
Hodgkins disease, 92, 93, 205, 247, 279,
302, 317 seealso lymphoma
homeless people, 192
homeopathic immunisation, 359
homosexual men see sexual contact
HPV infection see human papillomavirus
(HPV)
human diploid cell vaccine (HDCV), 42,
111, 117, 118, 119 seealso rabies
human immunoglobulin see
immunoglobulins
human papillomavirus (HPV), 59, 16474
adverse events, 1734
contraindications, 173
dosage and administration, 171
males, 173
precautions, 173
pregnancy, 88, 174
recommendations, 1713
human rabies immunoglobulin (HRIG),
112, 1147, 176 seealso rabies
hyperimmune globulin, 88, 102
hypotonic-hyporesponsive episode
(HHE), 233, 235
HZ see zoster

Index 385

Index

immigrants, 22, 75, 108


hepatitis B, 150
rubella, 278
immune thrombocytopenia, 180 seealso
bleeding disorders
Immunisation History Statements, 22, 68
immunocompromised see impaired
immunity, individuals with
immunodeficient see impaired immunity,
individuals with
immunoglobulins, 1023, 17583 seealso
normal human immunoglobulin
(NHIG)
adverse events,

intramuscular NHIG, 1789


intravenous NHIG, 180
availability, 176
botulism antitoxin, 181
cytomegalovirus immunoglobulin,
1812
dosage and administration, 177, 179
normal human immunoglobulin
(NHIG), 175, 1767, 179
prophylaxis, 1778
recommendations,
intramuscular NHIG, 1778
intravenous NHIG, 180
respiratory syncytial virus
immunoglobulin, 1823
specific diseases and, 175, 181
hepatitis A, 146, 177
hepatitis B, 151, 157, 163, 177
measles, 177, 210
rabies (HRIG), 112, 1147, 176
rubella, 280
smallpox, 2867
tetanus, 2945
varicella, 178, 317
zoster immunoglobulin (ZIG), 178,
309, 31920
immunosuppressed see impaired
immunity, individuals with
immunosuppressive agents, 118, 205,
279, 300, 317
Imogam Rabies, 112 seealso rabies
impaired immunity, individuals with,
902, 349
cholera, 122, 123
contacts of, 20, 91, 192, 209, 271, 285,
308, 313, 3145, 320, 321
Haemophilus influenzae type b, 132
hepatitis B, 158, 160, 161
human papillomavirus (HPV), 173
immunoglobulins, 178, 180
influenza, 90, 91, 92, 191
Japanese encephalitis (JE), 199
measles, 91, 205, 211
mumps, 91
pneumococcal disease, 901, 240, 247
rotavirus, 266, 271
rubella, 91, 27980
smallpox, 91

tuberculosis, 91, 298


typhoid, 91
varicella, 91, 309, 3167
yellow fever, 91, 326
zoster (herpes zoster) (HZ), 329
inactivated poliomyelitis vaccine (IPV),
42, 77, 2534 seealso poliomyelitis
inactivated vaccines, 878
cholera, 121
diphtheria-containing, 1256
dT, 126, 290
DTPa-containing, 1256, 152, 134,
253, 22930, 28991
dTpa-containing, 126, 254, 2901,
Haemophilus influenzae type
b-containing, 134
hepatitis A-containing, 141
human papillomavirus (HPV), 169
influenza, 1878
Japanese encephalitis (JE), 196
meningococcal, 215, 216
pertussis-containing, 22930
pneumococcal, 241, 242
poliomyelitis-containing, 2534
rabies, 111
typhoid, polysaccharide vaccine, 304
incomplete dose see interruption to a
vaccination; regurgitation of vaccine
India
poliomyelitis, 251, 252
typhoid, 304
Indigenous people see Aboriginal and
Torres Strait Islander people
Indonesia
JE virus, 198
poliomyelitis, 252
rabies, 119
Infanrix hexa, 42, 125, 134, 152, 230, 253,
289 seealso DTPa-hepB-IPV-Hib
Infanrix-IPV, 42, 125, 230, 254, 290 seealso
DTPa-IPV
Infanrix Penta, 42, 125, 152, 229, 253, 290
seealso DTPa-hepB-IPV
infants see children and infants
infectious period
hepatitis A, 146
measles, 201
pertussis, 2356

varicella, 309
inflammatory bowel disease, 61, 122, 209,
3523
influenza, 723, 77, 18494, 354
adverse events, 1934
contraindications, 193
dosage and administration, 18990
precautions, 193
pregnancy, 88, 192, 194
preterm babies, 90
recommendations, 1903
variation from product information,
194
Influvac, 42, 187 seealso influenza
injecting drug users, 109, 139, 144, 145,
158, 293
injection site nodules, 60, 356
seealso adverse events following
immunisation (AEFI)
injection site, 456, 515 seealso dosage
and administration; intradermal
injections; intramuscular injections;
subcutaneous injections
injection technique, 42, 445 seealso
dosage and administration;
intradermal injections; intramuscular
injections; subcutaneous injections
intellectually disabled people, 144, 145,
159
interchangeability of vaccines
Haemophilus influenzae type b, 31, 136
hepatitis A, 1412
hepatitis B, 161
poliomyelitis, oral and inactivated,
255
rotavirus, 273
interferon-gamma release assays, 302
interruption to a vaccination, 25 seealso
regurgitation of vaccine
intradermal injections, 1134
Intragam P, 179, 180 seealso
immunoglobulins
intramuscular injections, 42, 4455
intussusception (IS), 272, 362
invasive pneumococcal disease (IPD), 72,
2401, 246, 247
IPOL, 42, 253 seealso poliomyelitis

386 The Australian Immunisation Handbook 9th Edition

Japanese encephalitis (JE), 73, 78,


195200
adverse events, 199200
contraindications, 199
dosage and administration, 197
precautions, 199
pregnancy, 88, 200
recommendations, 1978
variation from product information,
200
jaundice, false contraindication, 21
JE-VAX, 42, 196 seealso Japanese
encephalitis (JE)

Kawasaki disease, 103, 179, 180

mad cow disease, 354 seealso bovine


spongiform encephalopathy (BSE)
Mantoux technique see tuberculin skin
test (TST)
manufacturing residuals, 3403, 357
maternity hospital staff, 105, 2378
Maternity Immunisation Allowance, 67,
68, 347
measles, 77, 105, 20112 seealso MMR
adverse events, 2089
catch-up, 32, 38
contraindications, 2056
dosage and administration, 203
immunoglobulin, 1778, 2102
precautions, 2068
pregnancy, 86, 212
public health management, 20912
recommendations, 2035
transmissibility, 209
variation from product information,
212
measles-mumps-rubella vaccine
see MMR
measles-mumps-rubella-varicella
vaccine see MMRV
meat industry, 106, 258, 260
medical students, 105, 145, 279
Medicare, 67, 332
men who have sex with men see sexual
contact
Mencevax ACWY, 42, 216, 222 seealso
meningococcal disease
Meningitec, 42, 215, 222 seealso
meningococcal disease
meningococcal disease, 79, 101, 21322
asplenia (anatomical or functional)
and, 101, 218, 219
early clinical management and, 220
meningococcal C conjugate vaccine
(MenCCV), 215
adverse events, 220

Index 387

Index

laboratory personnel, 106, 113, 198, 218,


219, 255, 260, 284, 306, 324
lactation see breastfeeding and lactation
latent TB infection (LTBI), 301
Latin America see Americas
leprosy, 298, 300
leukaemia, 16, 93, 94, 178, 205, 279, 285,
317, 349
limb swelling, 59, 234
Liquid PedvaxHIB, 42, 31, 71, 133, 134
seealso Haemophilus influenzae type b
live attenuated vaccines, 201, 85, 91
BCG, 298
contraindications and, 20, 85, 86, 91
immunoglobulins/blood products
and, 16, 18, 1023, 1756, 271
interval between, 17, 206, 225, 280,
301
MMR, 201, 223, 274
rotavirus, 267
smallpox, 284
surgery and, 104
typhoid (oral), 304
varicella, 309
yellow fever, 323
liver transplant recipients, 94, 144, 159
livestock industry, 107, 258, 260

local anaesthetic, vaccinations and


see topical anaesthetics
lockjaw, 288 seealso tetanus
lymphoma, 93, 205, 247, 279, 285, 302,
317 seealso Hodgkins disease

catch-up, 32
contraindications, 219
dosage and administration, 217
recommendations, 218
meningococcal polysaccharide
vaccine (4vMenPV), 215
adverse events, 220
contraindications, 219
dosage and administration, 217
recommendations, 219
pregnancy, 87, 2212
public health management, 2201
variation from product information,
222
Menjugate Syringe, 42, 215, 222 seealso
meningococcal disease
Menomune, 42, 216, 222 seealso
meningococcal disease
mercury, 355 seealso thiomersal-free
vaccines; thiomersal in vaccines
Mrieux Inactivated Rabies Vaccine, 42,
111 seealso Australian bat lyssavirus
(ABL) infection; rabies
Meruvax II, 42, 277 seealso rubella
Middle East seealso Hajj
cholera, 120
meningococcal disease, 219
military personnel see armed forces
minimum ages for vaccination, 30
minimum dose intervals for National
Immunisation Program (NIP) vaccines,
29
MMR (measles-mumps-rubella), 77,
3523, 3589
catch-up, 32
for measles, 202
for mumps, 224
for rubella, 275
immunoglobulin/blood products
interval, 94, 1023, 147, 1756,
2102
MMRV (measles-mumps-rubellavaricella), 2024, 206, 224, 310, 312
multiple injections, 24, 567, 345
multiple sclerosis (MS), 61, 1012, 163,
191, 352, 353, 370
mumps, 202, 2236 seealso MMR
adverse events, 225

catch-up, 32, 38
contraindications, 2056, 225
dosage and administration, 224
immunoglobulin, 225
precautions, 225
pregnancy, 86, 225
recommendations, 2245
variation from product information,
212, 226
Mycobacterium tuberculosis
see tuberculosis

National Centre for Immunisation


Research and Surveillance of Vaccine
Preventable Diseases (NCIRS), iii, iv,
13, 353, 359
National Immunisation Program (NIP),
127, 215, 228
components of vaccines used in,
3403
National Measles Control Campaign,
202, 274
National Meningococcal C Vaccination
Program, 215
needles, 44 seealso dosage and
administration
Neisseria meningitidis see meningococcal
disease
NeisVac-C, 42, 215, 222 seealso
meningococcal disease
neurological disease, 21, 90, 191, 195,
199200, 233, 348
newborn nurseries, 2378, 271
normal human immunoglobulin (NHIG),
1467, 175, 177, 176, 179
seealso immunoglobulins
Normal Immunoglobulin-VF (human)
(NHIG), 177 seealso immunoglobulins
nurses see healthcare workers (HCW)
nursing home residents, 188, 192
nursing home staff, 106, 192
nursing students, 105, 145, 279

occupational risks, 1047


hepatitis A, 144, 145, 147
hepatitis B, 15960, 161, 162, 163

388 The Australian Immunisation Handbook 9th Edition

Pacific Islands see Oceania


palivizumab (Synagis), 176, 1823 seealso
immunoglobulins

Papua New Guinea, 75, 78, 82, 198, 304


paracetamol, 58, 60, 207, 209, 318, 348
pertussis, 31, 87, 96, 989, 1057, 125, 127,
134, 22739, 3467, 358 seealso DTPacontaining vaccines
adverse events, 2345
contraindications, 233
dosage and administration, 231
minimum interval between dTpa
and dT, 233
precautions, 2334
pregnancy, 87, 238
public health management, 2358
recommendations, 2312
special considerations, 233
variation from product information,
2389
Pharmaceutical Benefits Advisory
Committee (PBAC), 334
phenol, 342, 356
phenoxyethanol, 342, 355
plague, 3
pneumococcal disease, 72, 24050
asplenia (functional or anatomical),
1001
invasive pneumococcal disease
(IPD), 72, 2401, 246, 247
pneumococcal conjugate vaccine
(7vPCV), 135, 2412, 2436
Aboriginal and Torres Strait
Islander children, 345, 72,
2401, 243, 245, 247
adverse events, 249
catch-up, 32, 346
contraindications, 248
dosage and administration, 243
impaired immunity, individuals
with, 901, 92
Infanrix hexa and, 134
recommendations, 2436
pneumococcal polysaccharide
vaccine, (23vPPV) 241, 242, 2468
Aboriginal and Torres Strait
Islander children, 72, 245, 247
Aboriginal and Torres Strait
Islander people, 73, 241, 246, 248
adverse events, 249
contraindications, 248

Index 389

Index

influenza, 1923
Japanese encephalitis (JE), 198
measles, 204
meningococcal disease, 2189, 221
pertussis, 232, 237, 346
poliomyelitis, 255
Q fever, 258, 260
rabies, 113
rubella, 279, 281
smallpox, 284
typhoid, 306, 308
varicella, 313, 3156, 321
yellow fever, 324
occupation health and safety, 39 seealso
standard precautions
Oceania (includes Pacific Islands)
cholera, 120
hepatitis B, 77, 150
poliomyelitis, 251
rabies, 110
rubella, 278
tuberculosis, 297
typhoid, 78, 304
yellow fever, 322
Octagam, 179 seealso immunoglobulins
older people
influenza, 77, 188, 190, 193, 346
pertussis, 19, 232, 346
pneumococcal disease, 77, 101, 246,
248
tetanus, 288
varicella 330
yellow fever, 326
zoster (herpes zoster) (HZ), 329, 330
oncology patients, 924 seealso impaired
immunity, individuals with
oral poliomyelitis vaccine (OPV), 601,
252, 254, 255, 302, 305
oral vaccines, 42
organ donors see blood and organ donors
outbreak control see public health
management
overseas travel see travellers

dosage and administration, 243


impaired immunity, individuals
with, 91
recommendations, 2468
revaccination with, 2478
pregnancy, 87, 249
preterm babies, 89, 246
travel and, 77
variation from product information,
250
pneumonia, 723, 190, 240, 242
Pneumovax 23, 42, 242 seealso
pneumococcal disease
police, 106, 159, 192
poliomyelitis, 2516 seealso DTPa-hepBIPV-Hib; dTpa-hepB-IPV, inactivated
poliomyelitis vaccine (IPV); oral
poliomyelitis vaccine (OPV); vaccinederived poliovirus (VDPV)
adverse events, 255
catch-up, 32
contraindications, 255
dosage and administration, 254
pregnancy, 88, 255
recommendations, 2545
variation from product information,
256
polysaccharide vaccines see
meningococcal disease; pneumococcal
disease; typhoid
positioning for vaccination, 4751
post-exposure treatment
Australian bat lyssavirus (ABL)
infection, 1147
hepatitis A, 1467
hepatitis B, 1612
immunoglobulins, 1778
measles, 20912
meningococcal disease, 2201
mumps, 225
pertussis, 2378
rabies, 112, 1147
varicella, 314, 3156
post-vaccination procedures, 5869, 377
seealso serology
poultry industry, 107, 192
precautions, 14, 40, 348, 3501
cholera, 123

human papillomavirus (HPV), 173


immunoglobulins, 178, 180
influenza, 193
Japanese encephalitis (JE), 199
measles, 2068
mumps, 225
pertussis, 2334
Q fever, 263
rotavirus, 2702
rubella, 280
smallpox, 285
tetanus, 295
tuberculosis, 301
typhoid, 307
varicella, 3178
yellow fever, 326
prednisolone, 92, 205, 207, 246, 317, 349
pre-exposure prophylaxis
Australian bat lyssavirus (ABL)
infection, 1124
immunoglobulins, 177
rabies, 78, 81, 82, 1124, 119
pregnancy and vaccination, 16, 1820,
8490, 350
BCG vaccine, 86, 3001
cholera vaccine, 87, 123
contacts, vaccination in, 18, 86, 89,
350
diphtheria-containing vaccine, 87
Haemophilus influenzae type b
vaccine, 87
hepatitis A vaccine, 88
hepatitis B vaccine, 88
human papillomavirus (HPV)
vaccine, 88, 174
immunoglobulins, 88
influenza vaccine, 88, 192, 194
Japanese encephalitis (JE) vaccine,
88, 200
measles-containing vaccine, 86, 206,
212
meningococcal vaccine, 87, 2212
mumps-containing vaccine, 86, 225
pertussis-containing vaccine, 87,
232, 346
pneumococcal vaccine, 87, 249
poliomyelitis, 88
Q fever vaccine, 87, 263

390 The Australian Immunisation Handbook 9th Edition

pertussis-containing vaccine, 2389


pneumococcal vaccine, 250
poliomyelitis vaccine, 256
Q fever vaccine, 264
rabies vaccine, 119
rotavirus vaccine, 273
rubella vaccine, 282
tetanus-containing vaccine, 296
typhoid vaccine, 308
varicella vaccine, 321
yellow fever vaccine, 328
products previously available, 339
products registered but not currently
available, 339
public health management
diphtheria, 1289
Haemophilus influenzae type b, 1378
hepatitis A, 1467
measles, 20912
meningococcal disease, 2201
pertussis, 2358
rubella, 281
typhoid, 308
varicella, 31920
purified chick embryo cell vaccine
(PCECV), 42, 111, 117, 118 seealso
rabies
Purified Protein Derivative (PPD), 3012
seealso tuberculosis

Q fever, 106, 107, 25764


adverse events, 263
contraindications, 262
dosage and administration, 260
precautions, 263
pregnancy, 87, 263
pre-vaccination testing, 259, 2602
recommendations, 2602
serology, 2602
skin testing, 2612
variation from product information,
264
Q fever fatigue syndrome (QFS), 257
Q Fever Management Program, 258
Q-VAX, 42, 259 seealso Q fever
Q-VAX Skin Test, 259 seealso Q fever

Index 391

Index

rabies vaccine, 88, 118


rotavirus vaccine, 86
rubella vaccine, 86, 278, 280, 281, 282
smallpox vaccine, 86, 285, 286
tetanus-containing vaccine, 87
typhoid vaccine, 86, 87, 307, 308
varicella vaccine, 86, 313, 316, 320,
321
yellow fever vaccine, 86, 326, 327,
328
preschool see childcare facilities
preservatives in vaccines, 3403, 3556
preterm babies, 18, 45, 89 seealso babies
(neonates)
catch-up and, 31
Haemophilus influenzae type b
vaccine, 90, 136
hepatitis B vaccine, 90, 157
influenza vaccine, 90
pneumococcal vaccine, 89, 246
poliomyelitis vaccine, 255
rotavirus vaccine, 270
vaccination and, 3456
pre-vaccination procedures, 1438, 375
pre-vaccination screening, 821, 60, 375
seealso serology
checklist, 16, 375
Q fever, 2602, 263
tuberculosis, 299, 3012
Prevenar, 42, 241 seealso pneumococcal
disease
Priorix, 42, 203 seealso MMR; measles,
mumps; rubella
prisoners and staff see correction facility
inmates; correction facility staff
product information, variations from
BCG vaccine, 302
diphtheria-containing vaccine,
12930
Haemophilus influenzae type b
vaccine, 138
hepatitis B vaccine, 163
influenza vaccine, 194
Japanese encephalitis (JE) vaccine,
200
measles-containing vaccine, 212
meningococcal vaccine, 222
mumps-containing vaccine, 212

rabies, 78, 106, 107, 1109


adverse events, 118
contraindications, 117
dosage and administration, 112
endemic, 110, 111, 113, 115, 117
in Indonesia, 119
post-exposure prophylaxis,
vaccine, 1147
rabies immunoglobulin (HRIG),
103, 1167
pre-exposure prophylaxis, 78, 81, 82,
1124
pregnancy, 88, 118
variation from product information,
119
Rabipur Inactivated Rabies Vaccine, 42,
111 seealso Australian bat lyssavirus
(ABL) infection; rabies
refrigerators, 912
refugees see immigrants
regurgitation of vaccine, 25, 272 seealso
interruption to a vaccination
renal disease see chronic conditions
respiratory disease see chronic conditions
respiratory syncytial virus (RSV), 176
RSV immunoglobulin (RSVIG),
1823
respiratory syncytial virus
immunoglobulin (RSVIG) see
respiratory syncytial virus (RSV)
Reye syndrome, 191, 207, 310, 318, 321
rheumatoid arthritis (RA), 1012
rifampicin, 1378, 221
Rotarix, 42, 266, 267, 268 seealso rotavirus
RotaShield, 272
RotaTeq, 42, 266, 267, 268 seealso
rotavirus
rotavirus, 26573
adverse events, 2723
catch-up, 32
dosage and administration, 268
pregnancy, 86, 272
recommendations, 26870
regurgitation of vaccine, 25, 272
variation from product information,
273
route of administration, 412

rubella, 105, 27482, 358 seealso MMR


adverse events, 2801
blood products and MMR, 1023, 206
contraindications, 27980
dosage and administration, 278
immunoglobulins, 2812
males, 279
precautions, 280
pregnancy, 86, 281
public health management, 281
recommendations, 2789
serological testing for, 2767
variation from product information,
282

salicylates see aspirin therapy


Salmonella Typhi see typhoid
Sandoglobulin NF liquid, 179 seealso
immunoglobulins
Saudi Arabia, 79, 219
screening see pre-vaccination screening;
serology
seizures or convulsions, history of, 191,
207, 249, 348 seealso febrile convulsion
serology
post-vaccination, 113, 116, 142, 160,
276, 278, 314
pre-vaccination, 84, 97, 108, 143,
2601, 2767, 314, 315
sexual contact
hepatitis A, 146
hepatitis B, 1501, 158, 160, 162
HPV infection, 165
men who have sex with men, 108,
139, 144, 145, 158
shearers, 107, 258, 260
shingles see zoster
simultaneous administration of vaccines,
567 seealso multiple injections
BCG vaccine, 301
cholera vaccine, 122
Haemophilus influenzae type b, 135
hepatitis A vaccine, 144
hepatitis A/hepatitis B vaccine, 145
hepatitis A/typhoid vaccine, 145
human papillomavirus (HPV)
vaccine, 171

392 The Australian Immunisation Handbook 9th Edition

systemic lupus erythematosus (SLE),


1012

tattooists, 107, 159


technical terms, glossary of, 3648
tetanus, 77, 28896, 358 seealso DT; dT;
DTPa; dTpa
adverse events, 2956
contraindications, 295
dosage and administration, 291
precautions, 295
pregnancy, 87, 88, 296
recommendations, 2923
tetanus-prone wounds, treatment of,
292, 2935
variation from product information,
296
Tetanus Immunoglobulin-VF (for
intravenous use), 295 seealso tetanus
Tetanus Immunoglobulin-VF (TIG),
2945 seealso tetanus
tetravalent meningococcal
polysaccharide vaccines
see meningococcal disease
Therapeutic Goods Administration, 284,
354
thimerosal see thiomersal-free vaccines;
thiomersal in vaccines
thiomersal-free vaccines, 153, 156, 157,
355
thiomersal in vaccines, 153, 188, 343, 355
thrombocytopenia, 2078
tick-borne encephalitis, 79
topical anaesthetics, 41, 43, 116
Torres Strait Islanders see Aboriginal and
Torres Strait Islander people
Torres Strait Islands and Far North
Queensland see Far North Queensland
and Torres Strait Islands
transmission, live vaccines and, 859, 91
measles, 209, 212, 349, 350
mumps, 225, 349, 350
rotavirus, 270, 271
rubella, 280, 349, 350
typhoid (oral), 307
varicella, 3145, 318, 349, 350

Index 393

Index

immunoglobulins/blood products
and, 103
inactivated vaccines and
immunoglobulins, 176
influenza vaccine, 190
meningococcal vaccine, 217, 222
MMR vaccine, 203, 224, 225, 278, 280
oral cholera and oral typhoid
vaccines, 122
pneumococcal vaccine, 243, 246
Rh (D) immunoglobulin (anti-D)
and, 176, 206, 279
rotavirus vaccine, 268
rubella vaccine, 278
typhoid, 305
varicella vaccine, 312, 317
yellow fever vaccine, 326
skin cleaning, 43
smallpox, 85, 91, 106, 2837
adverse events, 286
contraindications, 285
dosage and administration, 284
immunoglobulin, 286
precautions, 285
pregnancy, 86, 286
recommendations, 284
smokers
pneumococcal disease, 247, 248
pregnancy and, 84, 87
solid organ transplant (SOT) recipients,
946, 145
splenectomy, 16, 19, 36, 93, 1001, 136,
247 seealso asplenia (functional or
anatomical)
Stamaril, 42, 323 seealso yellow fever
standard precautions, 39, 105, 159, 270
stem cell transplants see haematopoietic
stem cell transplantation (HSCT)
steroids see corticosteroids
stockyard workers, 107, 258, 260
Streptococcus pneumoniae
see pneumococcal disease
subcutaneous injections, 42, 55
sudden infant death syndrome (SIDS),
61, 235, 354
surgery, 21, 104
Synagis (palivizumab), 176, 1823 seealso
respiratory syncytial virus (RSV)

transplants see haematopoietic stem cell


transplantation (HSCT); solid organ
transplant (SOT) recipients
transport, storage and handling of
vaccines seealso cold chain
BCG vaccine, 298
cholera vaccine, 121
diphtheria-containing vaccine, 127
Haemophilus influenzae type b
vaccine, 135
hepatitis A vaccine, 142
hepatitis B vaccine, 153
human papillomavirus (HPV)
vaccine, 171
immunoglobulins, 176
influenza vaccine, 189
Japanese encephalitis (JE) vaccine,
197
measles-containing vaccine, 203
meningococcal vaccine, 217
mumps-containing vaccine, 224
pertussis-containing vaccine, 231
pneumococcal vaccine, 2423
poliomyelitis vaccine, 254
Q fever vaccine, 259
rabies vaccine, 112
rotavirus vaccine, 267
rubella vaccine, 277
smallpox vaccine, 284
tetanus vaccine, 291
typhoid vaccine, 305
varicella vaccine, 3112
yellow fever vaccine, 323
travellers, 7583, 91, 100 seealso Africa;
Americas; armed forces, Asia; Europe;
Hajj; Middle East; Oceania
cholera, 78, 120, 122
diphtheria, 128
hepatitis A, 78, 88, 144
hepatitis A/typhoid, 145
hepatitis B, 77, 159
infections acquired by, 756
influenza, 77, 193
Japanese encephalitis (JE), 78, 1978
measles, 77, 2045, 358
meningococcal disease, 79, 219
MMR, 2045
pneumococcal disease, 77

poliomyelitis, 77, 88, 252, 255


rabies, 78, 88, 111, 113, 115, 117
tetanus, 77, 293
tick-borne encephalitis, 79
tuberculosis, 79, 300
typhoid, 78, 86, 91, 303, 305, 306
varicella, 77
yellow fever, 79, 86, 91, 323, 3245,
326, 327, 328
tuberculin skin test (TST), 207, 299, 300,
3012
live vaccines and, 302
tuberculosis (TB), 701, 79, 207, 297302
adverse events, 301
contraindications, 300
dosage and administration, 2989
precautions, 301
pregnancy, 86, 301
recommendations, 300
tuberculin skin test, 3012
variation from product information,
302
Tubersol, 3012 seealso tuberculin skin
test (TST)
Twinrix, 141 seealso hepatitis A (HAV);
hepatitis B (HBV)
Twinrix Junior, 141 seealso hepatitis A
(HAV); hepatitis B (HBV)
Typherix, 42, 304 seealso typhoid
Typhim Vi, 42, 304 seealso typhoid
typhoid, 78, 123, 3038
adverse events, 307
contraindications, 307
dosage and administration, 3056
hepatitis A/typhoid, 143, 145, 304
precautions, 307
pregnancy, 86, 87, 308
public health management, 308
recommendations, 306
variation from product information,
308

Vaccination, commonly asked questions,


34459
vaccine-associated paralytic
poliomyelitis (VAPP), 601, 252

394 The Australian Immunisation Handbook 9th Edition

ventrogluteal area, 45, 46, 48, 51, 534, 56,


57, 345 seealso injection site; injection
technique
veterinarians, students and staff, 107,
113, 260
Vibrio cholerae see cholera
Vivaxim, 42, 141, 304 seealso hepatitis A
(HAV); typhoid
Vivotif Oral, 42, 304 seealso typhoid

whooping cough see pertussis


women seealso breastfeeding and
lactation; pregnancy and vaccination
of child-bearing age and rubella
vaccination, 2757, 278, 279
World Health Organization (WHO), 22,
78, 79, 83, 108, 163, 202, 219, 251, 252,
297, 353

yellow fever, 75, 79, 106, 3228


adverse events, 327
contraindications, 91, 100, 3256
dosage and administration, 324
endemic countries, 324
International Certificate of
Vaccination against Yellow Fever,
323, 325
precautions, 326
pregnancy, 86, 327
recommendations, 3245
variation from product information,
328
yellow fever vaccination centres, 325

zoster (herpes zoster) (HZ), 309, 319,


32931
vaccine, 3301, 339
zoster immunoglobulin (ZIG), 178, 309,
315, 31920 seealso varicella
Zoster Immunoglobulin-VF (human),
31920 seealso varicella

Index 395

Index

vaccine-associated viscerotropic adverse


events, 327
vaccine-derived poliovirus (VDPV), 252
vaccines
components used in NIP, 3403
dates when available in Australia,
3724
failures, 136, 259, 3101, 3123
vaccinia immune globulin (VIG), 2867
seealso smallpox
vaccinia virus (smallpox), 85, 106, 283
seealso smallpox
VAQTA, 42, 141 seealso hepatitis A
(HAV)
VAQTA Paediatric/Adolescent
formulation, 42, 141, 146 seealso
hepatitis A (HAV)
varicella, 77, 98, 99, 104, 105, 207, 30921
adverse events, 60, 318
breakthrough varicella, 3101, 3123
catch-up, 32
contraindications, 91, 92, 96, 3167
dosage and administration, 44, 312
immunoglobulin/blood product
interval, 94, 1023, 147, 1756,
3201
precautions, 92, 94, 3178
pregnancy, 86, 320
public health management, 178,
31920
recommendations, 3126
serological testing for, 314
variation from product information,
321
varicella-zoster virus (VZV), 309, 329,
330 seealso zoster
Varilrix, 42, 311 seealso varicella
Varivax Refrigerated, 42, 3112, 330
seealso varicella
vasovagal episodes, 612
vastus lateralis, 45, 46, 47, 512, 345
seealso injection site; injection
technique
Vaxigrip, 42, 187 seealso influenza
Vaxigrip Junior, 42, 188, 189 seealso
influenza

396 The Australian Immunisation Handbook 9th Edition

You might also like