You are on page 1of 168

548

15

Neoplastic Diseases

­Introduction
Venugopal Nair

Summary The first section describes MD, a T‐cell lymphoma


induced in chickens by the highly cell‐associated Marek’s
Agents, Infections, and Disease.  This chapter refers to all the disease virus (MDV). Marek’s disease lesions consist of
diseases that have neoplasm or cancer as a common feature. CD4+ T‐cell lymphomas affecting a number visceral
The majority of these diseases are caused by herpesviruses organs and tissues, together with lymphoid cell infiltra-
or retroviruses, designated as Marek’s disease (MD), tion into the peripheral nerves resulting in paralytic
avian leukosis, and reticuloendotheliosis. The chapter symptoms. Recent advances in MD research have pro-
also describes a set of tumors of a non‐infectious etiology. vided significant insights into the molecular mechanisms
of the disease, further strengthening its significance as
Diagnosis.  Most of the neoplastic diseases can be an excellent biomedical model for T‐cell lymphomas
diagnosed by the characteristic pathological changes in (3–6). Marek’s disease has been controlled since the early
the affected tissues, but confirmed by virological, 1970s by use of conventional live attenuated antigeni-
serological, and molecular methods of diagnosis. cally related vaccines. During the last four‐and‐a‐half
decades, research on MD has provided a better under-
Intervention.  While diseases such as MD is controlled standing of the viral gene functions, molecular mecha-
mainly by the widespread use of vaccines, most of the nisms of the disease and factors affecting host genetic
retroviral diseases are controlled by eradication of the resistance (7). However, despite widespread use of vac-
virus from the infected flocks, supplemented by selection cines and development of new methods of vaccination,
for genetic resistance. MD still remains a major challenge to poultry health,
particularly from the continuing increase in virulence of
Neoplastic diseases of poultry comprise a variety of condi- MDV strains (8, 9), possibly contributed by the vaccines
tions possessing a single common denominator: neoplasia themselves (10). Furthermore, the incidence of MD in
involving one more of the cell types. Unlike in human med- other avian species such as turkeys and geese (11, 12)
icine where the vast majority of cancers are of non‐infec- demonstrate the increasing host range and economic
tious origin (1), most of the neoplastic diseases affecting significance. Clearly, in the absence of control measures
avian health have a viral etiology. Indeed, studies on avian including vaccine failures, MD is capable of causing dev-
oncogenic viruses have contributed immensely to our cur- astating losses in poultry (13, 14). As a disease occurring
rent understanding of a number of molecular mechanisms worldwide, with reports of vaccination breaks and prob-
of cancer (2). This chapter deals primarily with the three able emergence of more virulent pathotypes, MD contin-
most economically important virus‐induced transmissible ues to pose severe threats to the poultry industry, and
neoplastic diseases of poultry, namely: (1) the herpesvirus‐ developing strategies for its control remains one of the
induced Marek’s disease (MD); (2) retrovirus‐induced great challenges today (7). This section gives an up to
avian leukosis/sarcoma, and (3) reticuloendotheliosis. An date account of the scientific understanding of MD and
additional final section covering tumors of unknown etiol- its control strategies.
ogy is also included. Each of these neoplastic diseases or A second section describes a group of leukoses, sarco-
disease complexes are described in a separate section mas, and related neoplasms induced by a number of
because of its etiologic distinctness. closely related groups of avian retroviruses termed the

Diseases of Poultry, Fourteenth Edition. Editor-in-chief David E. Swayne.


© 2020 John Wiley & Sons, Inc. Published 2020 by John Wiley & Sons, Inc.
Chapter 15  Neoplastic Diseases 549

leukosis/sarcoma (L/S) viruses. The contributions of chickens, commercial layers and native breeds of chick-
these retroviruses to tumor virology, such as the land- ens are affected (21–23). The section on leukoses/sar-
mark study on transmissible tumors, the discovery of the coma viruses gives a comprehensive coverage of the
oncogenes and the reverse transcriptase, have been hugely recent advances in our understanding of the pathogenic
significant. The term leukosis is used because a leukemic mechanisms, diagnosis, and eradication methods.
blood picture is not always present during the course of Technological advances in genome editing and the
leukemia‐like proliferative diseases of the hemopoietic potential for inducing genetic resistance as a tool for dis-
system (15–17). The various forms of hemopoietic sys- ease control are also mentioned (24).
tem neoplasia induced by the L/S group of avian retrovi- The third section describes reticuloendotheliosis
ruses include the lymphoid leukosis (LL), myeloid (RE), a group of disease syndromes caused by reticu-
leukosis (ML), and erythroid leukosis (EL) caused by dif- loendotheliosis virus (REV), unrelated to the leukoses/
ferent subgroups of the virus. Primarily affecting the sarcoma group of viruses (25). The REV group includes
bursa of Fabricius and visceral organs, LL is the most different viruses associated with a number of disease
common form of leukosis although efforts in the past syndromes. These consist of defective REV‐T strain
have successfully eradicated these viruses from many of that induces rapid neoplastic transformation by virtue
the commercial breeding flocks (15, 18). Other neo- of the very potent virus‐encoded oncogene v‐Rel, an
plasms of hematopoietic origin that can also be seen in NFkB homolog (26). The most common clinical
avian leukosis virus (ALV)‐infected chickens, albeit ­diseases induced by REV include chronic lymphoma
infrequently, include erythroblastosis, myeloblastosis, as  well as immunosuppressive runting disease.
myelocytomatosis, and certain related neoplasms such Reticuloendotheliosis virus infects chickens, turkeys,
as nephroblastoma and osteopetrosis (19). With the ducks, geese, pheasants, quail, and probably many other
emergence of a new ALV subgroup J in the late 1980s, avian species. However, a major economic concern of
myelocytomatosis, became a major neoplasia particu- REV arises from its potential as contaminants of live
larly in meat‐type chickens (16, 20). The incidence of vaccines produced in chicken embryo cells or tissues.
ALV‐J has been significantly reduced in Europe and Reticuloendotheliosis virus could also be a barrier for
North America through the successful implementation the export of breeding stock to certain countries. The
of eradication programs. However, it is alarming that sig- section gives very detailed account of some of patho-
nificant losses continue to occur in countries such as genic mechanisms of REV, and the recent developments
China, where in addition to the commercial meat‐type in diagnosis and control.

Table 15.1  Transmissible neoplasms.

Virus type Nucleic acid type Virus classification of etiological agent Neoplastic diseases

Retrovirus RNA Leukosis/sarcoma group Leukoses


Lymphoid leukosis
Erythroblastosis
Myeoloblastosis
Sarcomas and other connective tissue tumors
Fibrosarcoma, fibroma
Myxosarcoma, myxoma
Osteogenic sarcoma, osteoma
Histiocytic sarcoma
Related neoplasms
Hemangioma
Nephroblastoma
Hepatocarcinoma
Osteopetrosis
Reticuloendotheliosis group Reticuloendotheliosis
Herpesvirus DNA Marek’s disease virus Marek’s disease
550 Section II  Viral Diseases

The fourth and final section describes tumors of problem. The dilemma is largely due to the fact that cer-
unknown etiology on the basis of morphologic charac- tain strains of these viruses induce some pathologic
teristics. Included are a wide variety of benign and malig- lesions difficult to distinguish from those induced by
nant neoplasms derived from muscle, epithelial, and another unrelated virus. The two major lymphoid neo-
nerve tissues; serous membranes; and pigmented cells. plastic diseases MD and lymphoid leukosis are particu-
Lymphoproliferative disease (LPD), another neoplastic larly confusing. Although REV‐induced lymphomas are
disease of turkeys reported in Europe and Israel is also infrequent and generally arise from contaminated vac-
induced by another retrovirus (27). The incidence of cines, it could also add to the problems in differential
LPD of turkeys has always been sporadic and hence not diagnosis. A practical and useful strategy for the differ-
included in this chapter. ential diagnosis of viral lymphomas has been proposed
Because many of the avian tumor viruses appear to (28). The choice of terminology for the neoplasia used in
have multipotent characteristics, that is, they can some- this chapter (see Table 15.1) is based on that originally
times induce a variety of neoplasms, classification and adopted by the World Veterinary Poultry Association
nomenclature of virus‐induced neoplasms present a (29) with suitable modifications.

­Marek’s Disease
Venugopal Nair, Isabel Gimeno, and John Dunn

Summary Because the literature on MD has greatly expanded, it


is no longer feasible to cite all relevant publications that
Agent and Disease.  Marek’s disease (MD) is widespread provide the scientific basis for our current knowledge of
disease of chickens characterized by rapid‐onset the disease. In this chapter, literature is cited selectively,
lymphoid tumors, immunosuppression, and paralysis and reviews are often substituted for original papers.
caused by the highly contagious Marek’s disease virus, an Readers are advised to refer to the chapter on MD from
alphaherpesvirus belonging to Mardivirus genus. the previous editions of this book for more details from
previous years. Useful recent books on MD are Marek’s
Diagnosis.  Marek’s disease virus is ubiquitous and Disease (259) and Marek’s disease: An evolving problem
detecting the virus alone is not sufficient to make a (158) and proceedings from the international symposia
confirmatory diagnosis, unless it is associated with on MD (1).
characteristic clinical signs and lesions including visceral
tumors and peripheral nerve lesions. A number of Definition and Synonyms
molecular diagnostic tests based on the detection of viral
The seminal description by Jozsef Marek (360) identified
nucleic acids and viral proteins.
the disease as polyneuritis. Other common synonyms
included neuritis, neurolymphomatosis gallinarum, and
Intervention.  Control of the disease is a challenge because range paralysis. Jungherr and colleagues (294) proposed
of the ubiquitous nature of the virus, latent infection, and that the term lymphomatosis be subdivided into visceral,
continuous shedding of the virus from the infected birds neural, and ocular forms. In 1961, Biggs (44, 48) p
­ roposed
and long‐term persistence of the virus outside the host in the term Marek’s disease to distinguish the condition
the poultry environment. Vaccination is the cornerstone of clearly from etiologically different lymphoproliferative
the control of MD along with improved biosecurity diseases.
measures. Marek’s disease has also been subdivided into acute
and classical forms, where the latter term designates
forms of the disease prevalent prior to the 1950s (44).
Introduction Marek’s disease virus (MDV) can also induce other clini-
cally distinct disease syndromes such as transient paral-
Marek’s disease (MD) is a common lymphoproliferative ysis, early mortality syndrome, cytolytic infection,
disease of chickens, usually characterized by mononu- atherosclerosis, and persistent neurological disease.
clear cellular infiltrates in peripheral nerves and various
other organs and tissues including iris and skin. The dis-
Economic Significance
ease is caused by a herpesvirus, is transmissible, and can
be distinguished etiologically from other lymphoid Prior to use of vaccines, MD constituted a serious eco-
­neoplasms of birds. nomic threat to the poultry industry causing up to 60%
Chapter 15  Neoplastic Diseases 551

mortality in layer flocks and 10% condemnations in Etiology


broiler flocks. Because vaccines are not 100% effective,
sporadic losses still occur, but they are no longer as seri- Classification
ous a problem. In 2004, the worldwide annual losses
from MD were in the range of US$1 to 2 billion, although Marek’s disease virus is a cell‐associated herpesvirus
the authors have indicated that these figures are impos- (citations in [484]) with lymphotropic properties similar
sible to verify (381). The disease remains a major con- to gammaherpesviruses. However, its molecular struc-
cern for the poultry industry due to the unpredictability ture and genomic organization are similar to alphaher-
of outbreaks and the possibility that vaccines may ulti- pesviruses (68, 335, 567). As per the recent classification
mately fail as a consequence of the evolution of more by the International Committee on Taxonomy of
virulent strains of MDV. Viruses (ICTV) all MDV serotypes are grouped together
in the genus Mardivirus (314). Members of the genus
Mardivirus, described previously as three serotypes are
Public Health Significance now grouped as three species: Gallid alphaherpesvirus 2
Although there are several reports suggesting a role for (serotype 1), Gallid alphaherpesvirus 3 (serotype 2), and
MDV in the etiology of multiple sclerosis, there has Meleagrid alphaherpesvirus 1 (herpesvirus of turkey
been no evidence to suggest the MDV is infectious to [HVT], serotype 3). Serotype 1 MDV is the prototype
humans (496, 562) or associated with human cancer virus for this group of avian viruses, and except where
(455). otherwise indicated MDV refers to serotype 1 virus. On
the basis of their virulence, serotype 1 strains are further
divided into pathotypes, which are often referred to as
Scientific Significance mild (m)MDV, virulent (v)MDV, very virulent (vv)MDV,
Research on MD has contributed greatly to veterinary and very virulent plus (vv+)MDV strains (597, 604).
medicine, basic science, and comparative oncology. The Nononcogenic Gallid herpesvirus 2 isolated from chick-
disease is uncommonly complex, featuring an interplay ens (50, 125, 490) and HVT (307, 613) belonging to genus
of neoplasia and inflammation expressed as several dis- Mardivirus are also included in this chapter.
tinct clinical syndromes, each modified in important
ways by host genetic influences. Marek’s disease virus, Morphology
an alphaherpesvirus with lymphotropic properties of
gammaherpesviruses, is highly cell‐associated but read- The morphology and morphogenesis of MDV have been
ily transmitted, and its virulence varies and evolves. It reviewed (142, 163, 164). Hexagonal nucleocapsids
has two unique sister viruses, both nononcogenic, that 85–100 nm in diameter and enveloped particles 150–
naturally infect chickens and turkeys. Infection induces 160 nm in diameter may be seen in thin sections of
complex immune responses usually resulting in high infected cell cultures. Keratinocytes derived from
levels of protection. Vaccination for MD constitutes an chicken embryonic stem‐cells showed capsids/virions
outstanding example of successful disease control in although extracellular viruses could not be demonstrated
veterinary medicine, and MD vaccines are the first (141). Enveloped virus particles appearing as irregular
effective vaccines against cancer in any species. amorphous structures and measuring 273–400 nm have
Evolution of MDV virulence has also been shown as an been demonstrated in negatively stained preparations of
example of a biological arms race of pathogens in the feather follicle epithelium (FFE) (92). Thin‐section prep-
face of vaccination (57). arations of the FFE revealed large numbers of cytoplas-
mic enveloped herpesvirus particles in keratinizing cells.
The morphology of MD virions in cell cultures and FFE
History is shown in Figure 15.1A.
The morphology of serotype 2 and 3 strains resembles
The seminal report by József Marek, published in 1907 that of MDV serotype 1 (393, 427, 490). In thin sections,
(360), of paresis in four roosters is the first account of the however, nucleocapsids of HVT commonly show a
disease named after him. A detailed history of MD unique crossed appearance (393).
research can be found in the previous edition of Diseases
of Poultry (499), a paper by Professor Biggs (47), the vide-
Chemical Composition
otape Legacy of the 1960s, and the historical archives of
the American Association of Avian Pathologists. A long Viral DNA
view on the last 40 years of MD research has recently Physical Properties.  The complete sequence of several
been published as part of the 40th Anniversary of Avian strains belonging to the three serotypes confirmed
Pathology (51). that  the genomes are very similar consisting of linear,
552 Section II  Viral Diseases

(A) (B)

(C)

Figure 15.1  Electron micrographs of Marek’s disease virus (MDV). (A) Thin section of cultured duck embryo fibroblasts infected with MDV
showing scattered virions in nucleus. ×38,400. (B) Thin section of cultured duck embryo fibroblast infected with MDV, showing enveloped
virions in a nuclear vesicle. ×360,000. (C) Thin section of feather follicle epithelium (FFE) of chicken infected with MDV showing enveloped
virions within the cytoplasmic inclusions. Note difference in morphology compared with (B). ×70,000. (413). (Nazerian)

double‐stranded DNA molecules of approximately 160– Structural Organization.  The genomic structure of the three
180 kb with a buoyant density in neutral CsCl of 1.706 g/ serotypes is typical for alphaherpesviruses as previously
mL for serotype 1 (158, 499). It is difficult to separate suggested (114) with a unique long (UL) and a unique short
viral DNA from host cell DNA because its density is (US) sequence. These unique sequences are flanked by sets
close to that of chicken DNA. Cloning of the complete of inverted repeat sequences: the terminal repeat long
MDV genome as bacterial artificial chromosomes (BAC) (TRL), internal repeat long (IRL), internal repeat short
has greatly facilitated the production of MDV DNA (506, (IRS), and terminal repeat short (TRS), respectively. Alpha
645). Infectious BAC clones of a number of strains have (a)‐like sequences, believed to be important for the cleavage
been constructed allowing rapid manipulation of MDV and packaging of viral DNA into virions, are located at the
genomes to identify various determinants associated terminal ends of the TRL and IRL and between the IRL and
with MD biology (118, 313, 541). Similarly, the rescue of IRS regions (11, 282, 335, 540, 567).
infectious MDV from overlapping cosmid clones of the The complete genome sequences of a number of sero-
Md5 strain of MDV has also been reported (463). type 1 MDV strains as well as other vaccine serotypes
Chapter 15  Neoplastic Diseases 553

have now been determined (499). Comprehensive analy- passage and attenuation in serotype 1 strains (204, 260,
sis of the sequence of the TRL/IRL regions in the 528). This expansion was caused by a tandem amplifica-
genomes of 13 strains of varying virulence has identified tion of direct 132 bp repeat (266, 359, 476). Other
several single nucleotide polymorphisms (SNPs) which changes have also been described including the deletion
loosely partition between attenuated and non‐attenu- of 400 bp in the Bam‐HI A fragment of CVI988 clone C
ated strains (537, 539). (988C) and 988 C/R6 (266) and a deletion of 200 bp in the
BamH1 L fragment of the vvMDV strain Md11 (587).
DNA Structure in  Infected Cells.  The structure of viral Additional changes have been reported for CVI988 in
DNA in infected cells is dependent on the virus–cell the meq gene (335, 538) and the ICP4 promoter/
interaction. Linear viral DNA can be found in nuclei of enhancer region of CVI988 (299). It is not clear whether
cells undergoing virus replication (114). It remains these differences are a consequence of cell culture pas-
unknown how viral DNA is maintained in latently sage or reflect strain differences.
infected, nontransformed cells (80, 376), although there
was some evidence on the role of epigenetic regulation of Viral Genes and Proteins
the viral genome (63). The status of DNA in transformed Over the last 25 years, a number of individual genes
cells has been difficult to determine in part because a of  MDV‐1 have been identified and sequenced, and
variable percentage of the transformed cells may undergo the  proteins have been characterized (478, 498).
viral replication at any point in time, in which case linear Comprehensive reviews based on the complete sequences
DNA can be detected. Initial investigations into the for the three serotypes have been published including
status of the MDV genome have indicated an absence of lists of open reading frames (ORFs) and their putative
integration (553), MDV genome is now thought to exist products (499). Table  15.2 summarizes the location of
as a mixture of integrated and episomal DNA (499). the ORFs and indicates the number of ORFs with homo-
Marek’s disease virus can be found integrated at multiple logues to HSV, the number of ORFs with homologues
sites in the chromosomes of cells derived from MDV‐ shared among the three serotypes, and the number of
related T‐cell lymphomas, suggesting a correlation unique genes for each serotype. Many of the genes in the
between integration and oncogenicity. The integration UL and the US regions have homologues with HSV and
sites are preferentially located near the ends of the equine herpesvirus‐1 and 4‐, and the genome organiza-
chromosomes within the telomeric region. The telomere‐ tion is similar to these two alphaherpesviruses (354). For
like sequences located at the terminal ends of the MDV this chapter, the MDV genes are grouped into two
genome are thought to assist in the preferential ­general categories: genes with homologues in alphaher-
integration of the viral DNA (312). Interestingly, MDV pesviruses and genes unique for MDV. Only the genes
encodes an RNA telomerase subunit (viral TR [vTR]) that are important for the pathogenesis and immune
that shares 88% sequence identity with the chicken TR responses will be reviewed briefly. The reader is referred
(cTR) gene (201). The role of telomeres and telomerases to the contemporary literature for additional informa-
in MDV integration, pathogenesis and oncogenesis has tion (158, 499).
been recently reviewed (312).
Genes with  Homologs in  Alphaherpesviruses.  This broad
Structural Changes by Recombination and/or Mutation.  category of genes can be divided into immediate early
Serotype 1 strains quickly develop altered biologic (IE), early, and late genes, which are with few exceptions
characteristics upon serial passage in vitro, such as loss of important for virus replication.
oncogenicity, reduced expression of glycoprotein C (gC)
(131), and decreased replication in vivo (494) indicating Immediate Early and  Early Genes with  Homology to  HSV. 
that spontaneous mutations may have occurred. The IE genes are important transcriptional regulators.
Accumulations of such mutations after continuous cell Four IE genes have been identified: intracellular protein
culture passages and generations of mixed population of (ICP)4, ICP0, ICP22, and ICP27. Anderson et  al. (15)
viruses have been recently demonstrated by sequence identified ICP4 as a 4245 bp ORF, but sequence data
analysis (537, 539). The gradual evolution of pathotypes indicated the presence of an ORF of 6969 bp. This agrees
toward greater virulence and the changes in biologic with the finding that two functional promoter/enhancer
properties of MDV during in vivo backpassage (591) regions are located upstream of the larger ORF and that
further support the mutability of MDV. the putative promoter/enhancer region for the short
These biological changes are accompanied by several ORF was nonfunctional in vitro (299). Proof that ICP4
molecular changes, although it is not clear which molec- protein is a transactivator was provided by transfection
ular change correlates with a specific biological change. of the MD cell line (MDCC) MSB‐1 with the short form
An expansion was found within the Bam‐HI D and H of ICP4, showing increased transcription of the endoge-
fragments that are commonly associated with cell ­culture nous ICP4, pp38, and pp24 genes (185, 447).
554 Section II  Viral Diseases

Table 15.2  Number of tentative genes in the three serotypes of Marek’s disease virus (MDV) in relation to other alphaherpesviruses.a

Location of expected functional open reading frames (ORFs)b

Serotype Gene classification TRL (R‐LORF) UL (L‐ORF) IRL (R‐LORF) IRS (RS) US (S‐ORF) TRS(RS) Totalc

1 HSV Homologd 0 57 0 1 7 1 65,66


MDV‐specifice 1 4 1 0 1 0 6,7
h H
Serotype‐specific 13 8 13 2 3 2 26,41
Total 14 69 14 3 11 3 97,114
2 HSV Homolog 0 59 0 1 7 1 67,68
MDV‐specificf 1 4 1 0 1 0 6,7
Serotype‐specific 9 4 9 1 4 1 17,27
Total 10 66 10 2 12 2 90,102
3 HSV Homolog 0 59 0 1 8i 1 68,69
g
MDV‐specific 0 6 0 0 1 0 7,7
Serotype‐specific 4 2 4 6 1 6 13,23
Total 4 67 4 7 10i 7 88,99
a
 For reference see (15, 279, 344, 373).
b
 Based on the location of the start codon.
c
 The italic numbers indicate the number of single genes for each serotype; the bold figures give the total number of genes including the
duplications in the repeat regions.
d
 Based on the sequence of the GA strain, nomenclature adapted from (340).
e
 Serotype‐specific genes with homologues present in serotype 2 or 3.
f
 Serotype‐specific genes with homologues present in serotype 1 or 3.
g
 Serotype‐specific genes with homologues present in serotype 1 or 2.
h
 The sequence for Md5 has minor differences compared with GA.
i
 Includes 2 copies of US8.

The MDV ICP27 phosphoprotein (465) localizes in the shown to be sufficient in inducing protection against viru-
nucleus, can transactivate pp38 and pp14 independently lent MDV challenge (28). Similarly, some of the peptides
of ICP4, and represses the early thymidine kinase gene derived from gB showed antiviral properties providing
(466). Additionally, MDV ICP27 also interacts with SR some insights into MDV entry into cells (583).
proteins and inhibits splicing of cellular telomerase The UL44 gene encodes gC, a 57–65 kDa glycoprotein
chTERT and viral vIL8 transcripts (14). ICP0 (LORF1) identified in some early references as gA, which is exten-
has been identified as an ORF in the TRL and IRL, and a sively synthesized in productively infected cells and is
recent study using proteomic approaches has demon- expressed on the cell surface. In addition, gC is actively
strated that the ICP0 gene product is expressed in MDV‐ secreted by infected cells (139, 272–274) and is one of
infected CEF (348). the major antigens to which the chicken immune system
mounts a substantial serological response. More recently,
Late Genes.  The late gene products include the nucle- gC and UL13 were shown to be important in horizontal
ocapsid proteins, the tegument proteins, including VP16, transmission (286).
and the glycoproteins (354). The glycoproteins (gB, gC, The importance of gD, coded by US6, is poorly under-
gD, gE, gH, gI, gK, gL, and gM) are presumed to be stood. It is nonessential for horizontal transmission (16)
important for infection of cells, transfer of virus from cell and is expressed poorly (406, 552) in vitro probably as a
to cell, and immune responses. consequence of no or limited transcription. Limited
gB, encoded by UL27, consists of a complex of three gly- expression of gD compared to pp38 and gB has been
coproteins with molecular weights of 100, 60, and 49 kDa described in FFE (398), suggesting that specific tran-
and is important for cell attachment and/or ­penetration scription factors in the FFE may be needed for the pro-
based on the production of gB‐specific virus‐neutralizing duction of gD.
(VN) antibodies (reviewed by [354]). Deletion of gB from The functions of the other glycoproteins have not been
MDV prevented the cell to cell spread demonstrating the studied in detail. The gI and gE proteins interact with each
essential nature of this protein for MDV replication (506). other based on immunoprecipitation assays (552).
In a recent study adenovirus‐based expression of gB was Associations of mutations in the gL with MDV pathogenesis
Chapter 15  Neoplastic Diseases 555

and reduced immune responses have been reported (479, lymphoma formation through targeted recruitment of B
555). Mutants constructed in a BAC clone carrying d ­ eletions cells and CD4+ CD25+ T cells (186).
in the gM, gI, or gE gene indicate that the encoded glycopro-
teins are essential for virus replication, because the deletion Viral Lipase.  All the three serotypes of MDV encode the
mutants are unable to transfer infectivity from infected to viral lipase gene (vLIP) (11, 282, 316, 335, 567). vLIP, a
uninfected cells (507, 564). soluble, glycosylated protein, is encoded by the LORF‐2
gene consisting of two exons. The first exon codes for the
Genes Unique for  MDV.  Several genes have been signal peptide, and the second exon codes for the lipase
identified that are unique for MDV strains (Table 15.2). activity. vLIP is probably an IE or early protein (296). The
Some of these genes are present only in serotype 1, and others glycosylated protein is required for the efficient lytic rep-
may have homologues in MDV serotype 2 and/or HVT. lication in birds (297).

Latency Associated Transcripts (LATs).  The LATs are a pp38/pp24.  The MDV phosphorylated protein com-
group of transcripts antisense to ICP4 and have been plex, often referred to as pp38/pp24, is coded by two
reviewed in detail (80, 376). These include a large 10 kb genes located at opposite ends of the UL region (654).
transcript as well as several spliced transcripts referred Details of the previous research to understand the role of
to as MSR (MDV small RNA) or SAR (small antisense pp38/pp24 have been reviewed in previous editions of
RNA). The importance of LATs for latency or transfor- the book (499). Homologs for pp38 have been identified
mation is unclear. Latency associated transcripts are in serotype 2 strains (282, 407) and HVT (11, 530), but
expressed in both lytically infected and transformed their functional relationships are not known.
cells, including the MDV positive QT35 cell line (633). The function of the pp24/pp38 complex has not been
elucidated. Originally, it had been linked to oncogenicity
Meq (Marek’s EcoQ).  The molecular biology of Meq (R‐ because pp38 is expressed in the cytoplasm of a variable
LORF7) has been reviewed (324, 376, 388, 416, 551). The proportion of MDV‐transformed, latently infected lym-
Meq protein of 339 amino acids contains a basic leucine phocytes (146, 271). Expression of pp24/pp38 can be
zipper (bZIP) domain at the N terminal closely resembling enhanced by activation (447, 633) suggesting that pp38
the jun/fos oncogene family. Several studies, inclusion may play a role during reactivation and subsequent virus
deletion analysis have clearly demonstrated the critical replication. pp38 is essential for cytolytic infection of B
role of Meq in oncogenesis (352). Meq gene also shows cells and maintenance of transformed state (463),
diversity among different isolates and it has been sug- although deletion did not affect the ability of the virus to
gested that the positive selection may be driving evolution spread horizontally (228). CVI988 also expresses pp38,
(410, 626). Variations in the sequence of the proline‐rich but shows an amino acid mutation in an epitope defined
domains also showed association with virulence (512). by monoclonal antibody (mAb) H19 (148). The demon-
Meq shows differential binding to different promoters stration that vvMDV5 strain expressing the pp38 protein
depending on its dimerization status. As homodimers or from CVI988 remains oncogenic indicates that the
heterodimers with leucine zipper proteins, Meq can trans- attenuation of CVI988 is not associated with pp38 (329).
activate different promoters (64, 546, 547) resulting in the
upregulation of a number of genes including interleukin The 1.8 kb Gene Family.  The promoter/enhancer regions
(IL)‐2 and CD30, a member of the tumor necrosis factor of pp38 and pp24 are part of a bidirectional promoter
receptor II (TNFR‐II) family (78, 85). A number of subse- complex regulating the transcription of pp38/pp24, the
quent studies have demonstrated efficacy of Meq‐deleted 1.8 kb gene family and the origin of replication (165, 522).
viruses as efficient vaccines (331, 336, 526). The mecha- Please refer to the chapter in the previous edition of this
nism behind such improved immunogenicity is not fully book for detailed descriptions of the previous research on
understood, however gene expression changes in Meq‐ this gene family (499). Several IE transcripts originate
deleted viruses (336) and reduced immunosuppressive from the 1.8 kb gene family containing three exons (376,
effects (344) are thought to contribute to this effect. 478). These transcripts are truncated in attenuated strains
due to an expansion of a tandem 132 bp direct repeat (DR)
vIL‐8.  The vIL‐8 gene (R‐LORF2) is located in the long (132 bp DR) (58, 476). The difference in the numbers of
repeat region and originally was identified as a spliced copies of these repeats between virulent and vaccine
meq variant (432, 433). The gene consists of three exons strains forms the basis of their differentiation by polymer-
and is expressed late during cytolytic infection. IL‐8 ase chain reaction (PCR) assays (34, 525, 655), although
attracts T cells, especially after IL‐8 receptors are upreg- these are not directly linked to the oncogenicity (527).
ulated by interferon‐γ (IFN‐γ) suggesting a role in the
switch of infection from B to T lymphocytes (502). Telomerase RNA (vTR).  The existence of a unique gene
Subsequent studies have also shown that vIL‐8 promotes encoding the RNA telomerase subunit (vTR) was identified
556 Section II  Viral Diseases

in the IRL/TRL region of the MDV genome (201). Marek’s data demonstrate the role of the US3‐encoded kinase in
disease virus vTR showed nearly 88% sequence identity to the morphogenesis as well as cell to cell spread of virions
the chicken telomerase RNA (ChTR) indicating its trans- through the effect on stress fiber breakdown and polym-
duction from the host genome. vTR, regulated also by the erization of actin (509). The glycoproteins gE, gI, and gM
action of c‐myc (523), can constitute telomerase activity by play a role in the transfer of virus from infected to unin-
interacting with chicken telomerase reverse transcriptase fected cells (507, 564). Replication rates vary with sero-
(ChTERT) more efficiently than ChTR (201, 202). The type, passage level of the virus strain, cell type, and
direct association between MDV oncogenicity and vTR was temperature of incubation.
demonstrated using MDV lacking either one or both copies The spread of virus in vivo from cell to cell will require
of vTR (119, 565) that resulted in significantly impaired intimate contact between infected and uninfected cells,
ability to induce lymphomas with reduced ability for dis- which are most often lymphocytes, although epithelial
semination (303–305). The role of telomeres and telomer- cells also can be involved in this process. The precise
ase in MD pathogenesis has recently been reviewed (312). interaction between these cells remains one of the
important unsolved issues, although recent demonstra-
MDV‐encoded microRNAs.  MicroRNAs (miRNAs) are a tion of epithelial‐specific expression of the UL47
distinct class of small regulatory molecules of approxi- ­tegument protein (287) suggest distinct virus–host inter-
mately 22 nucleotides affecting gene expression in vari- actions in these cell types.
ous cell types (635). These have been identified in a large
range of organisms including several herpesviruses (389). Virus‐cell interactions
A number of miRNAs have been identified in MDV many Three general types of virus–cell interactions are recog-
of which are expressed at very high levels (138, 561, 636, nized: productive, latent, and transforming.
639). Some of these miRNAs have been shown to have
direct roles in pathogenesis (556, 652, 653, 656). Productive Infection.  During productive infection,
replication of viral DNA occurs; proteins are synthesized,
Other Unique Genes.  Proteins have not been identified and in some cases, virus particles are produced.
for several unique ORFs that are transcribed in tumor Replication is correlated with virulence (172), but with
cells. Most of these have not been further studied with a all serotypes the number of genome copies per cell can
few exceptions. RLOR5a (400) is expressed in tumor cell increase 100‐fold and exceed 1,200 in the case of HVT
lines, although its function remains unknown. Similarly, (300). Two types of productive infection exist. Fully
RLORF4 has also been shown to be associated with productive infection in the FFE of chickens results in
tumor development (289). Marek’s disease virus also development of large numbers of enveloped, fully
encodes the ubiquitin‐specific protease domain within infectious virions (92). In productive‐restrictive
the major tegument protein‐coding gene UL36 (285, infection, most of the virions are nonenveloped and
570). Expressed at high levels in the FFE cells, it may have noninfectious. A variable number of the virions in
a role in virus morphogenesis in the feather (286). cultured cells may be enveloped, which can be recovered
as cell‐free, infectious virus by disruption of cells. In all
Viral Vectors.  Several nonessential sites in the three susceptible cells, productive infection leads to
serotypes of MDV can be used for the insertion and intranuclear inclusion body formation and lysis of the
expression of foreign and specific MDV genes (reviewed cell. A gene for the viral host shut‐off protein has been
in [261, 349, 412]). The anticipated advantages of MDV‐ identified, UL41 (354), that is probably responsible for
vectored vaccines are that these vaccines will protect the initiation of the lytic process. Lytic infection in vivo
simultaneously against MD and other pathogens, and can cause frank necrobiotic lesion formation. Because of
reactivation from latency will reinforce immune responses this, productive infection has been termed cytolytic, and
against MD and the other pathogens (18, 67, 584). the terms are used synonymously (89).

Latent Infection.  Latency and tumorigenesis in MD have


Virus Replication
been reviewed (387). Latent herpesvirus infections have
Replication of the three serotypes is typical of other cell‐ been defined as the presence of viral DNA in the absence
associated herpesviruses and has been reviewed exten- of viral transcripts and proteins, although LATs have
sively (47, 301, 473, 484, 498). For initial infection of been described for many herpesviruses. This definition
cultures or chickens by cell‐free virus, enveloped virions is appropriate for the nontransforming serotype 2 and 3
bind to cellular receptors probably by gB perhaps in strains. For serotype 1 strains, the distinction between
combination with other glycoproteins. Heparan sulfate, latency and transformation is often problematic. In both
a member of the glycosaminoglycans, has been identified cases, the viral genome is present, but no information is
as one of the cellular receptor molecules (338). Recent available on differences in transcriptional regulation
Chapter 15  Neoplastic Diseases 557

between latently infected and transformed cells, cultures (high‐passage virus). Small quantities of low‐
because  it is impossible to separate latently infected, passage virus can be obtained from infected cell cultures
nontransformed cells from noninfected cells. As a by lysing cells in SPGA (sucrose–phosphate–glutamate–
consequence, studies on latency have often been done in albumin) buffer (98). The production of cell‐free HVT is
MD transformed cell lines. best achieved by lysing heavily infected cell cultures. Cell‐
Marek’s disease virus latency mostly is associated with free MDV and HVT can be stored at −70°C or lyophilized
CD4+ T cells, although CD8+ T cells and B cells can also (98). Potency of both cell‐associated and cell‐free vac-
be latently infected (105, 337, 386). Fewer than five cop- cines can be affected adversely by storage temperature,
ies of the viral genome are present in latently infected reconstitution technique, choice of diluent, and holding
cells (473). Marek’s disease virus latency is maintained time and temperature after reconstitution (239, 423).
through various mechanisms including non‐random de
novo DNA methylation and histone modifications, with
Susceptibility to Chemical and Physical Agents
different genomic regions separated by chromatin
boundary elements (63, 351). The MDV genome can be The stability of cell‐associated MDV serotype 1 and 2
reactivated from latently infected cells and tumor cells by strains is completely dependent on the viability of the
inoculation of susceptible chickens, co‐cultivation with cells. Any treatment affecting cell viability will impact
permissive cells, and in vitro cultivation of latently directly the infectivity of virus stocks.
infected lymphocytes. The latter approach can be used Cell‐free MDV obtained from the skin of infected
to estimate the number of latently infected cells by enu- chickens was inactivated when treated for 10 minutes
meration of antigen‐positive cells at 0 hours and 48 hours at pH 3 or 11 and stored for 2 weeks at 4°C, 4 days at
in culture (107). 25°C, 18 hours at 37°C, 30 minutes at 56°C, or 10 min-
utes at 60°C (91). Dander, litter, and feathers from
Transforming Infection.  Transforming infections occur infected chickens are infectious and presumably con-
only in cells infected with serotype 1 MDV. Selection of tain cell‐free virus from the FFE bound to cellular
transformed cells from the background of immunologically debris. The infectivity of such materials was retained
committed and noncommitted cells (431) would facilitate for 4–8 months at room temperature (263, 602) and for
comparative studies on transformed cells in tumors and at least 10 years at 4°C (87). Virus infectivity was inac-
tumor cell lines. The search for specific surface markers tivated by a variety of common chemical disinfectants
associated with tumors has identified antigens, broadly within a 10‐minute treatment period (97, 262). Survival
referred to as MD tumor‐associated surface antigen of virus in litter may be affected adversely by increased
(MATSA), detected on cells from MD lymphomas and humidity (602).
lymphoblastoid cell lines but not on the surface of
productively infected cells (443, 623). Recent studies
Strain Classification
confirm that CD30hi expression is characteristic of MD
lymphomas suggesting that CD30 is a component of a Serotypes
critical intracellular signaling pathway perturbed in Following full genome sequencing, the three MDV
neoplastic transformation (78, 511). Both MATSA and serotypes have now been designated as three distinct
CD30 can be used to enrich for transformed cells in species: Gallid alphaherpesvirus 2 (serotype 1), Gallid
tumor cell suspensions. alphaherpesvirus 3 (serotype 2), and HVT (serotype 3).
Von Bülow and Biggs (576, 577) originally classified the
Virus Stock Production and Stability.  Productively infected MDV herpesvirus group into three distinct serotypes
cell cultures are a common source of cell‐associated that correlated with biologic properties. Type‐specific
virus stocks for all three viral serotypes and for cell‐free mAbs (270, 332) usually are used to determine virus
HVT stocks. Techniques for the production and serotype.
cryopreservation of cell‐free and cell‐associated virus A number of biological characteristics are associated
stocks have been described (reviewed in 130). Cell‐ with viral serotypes (50, 484). Low‐passage serotype 1
associated stocks of MDV or HVT are routinely stored at viruses grow best in duck embryo fibroblast (DEF) or
−196°C. The infectivity of such stocks, however, is chicken kidney cell (CKC) cultures, grow slowly, and
directly related to viability of the cells contained in these produce small plaques. Serotype 2 viruses grow best in
preparations and depends also on optimal freezing and chicken embryo fibroblasts (CEF), grow slowly, and pro-
thawing techniques. Under ideal conditions, the half‐life duce medium plaques with some large syncytia.
of diluted, cell‐associated virus stocks or vaccines should Herpesviruses of turkey grow best in CEF, grow rapidly,
be at least 2–6 hours (559). and produce large plaques. More infectious virus can be
Cell‐free serotypes 1 and 2 virus stocks are best extracted from HVT‐infected cells than from cells
obtained from FFE (low‐passage virus) or infected cell infected with serotype 1 or 2 viruses.
558 Section II  Viral Diseases

Pathotypes Attenuated MDV and serotype 2 and 3 viruses can be


Virulence or oncogenicity is only associated with sero- isolated readily and propagated in CEF (490). Infected
type 1 MDVs. Within this group, however, a wide varia- cultures usually develop discrete focal lesions, called foci
tion in pathogenic potential is recognized and or plaques, which consist of clusters of rounded, refrac-
undoubtedly represents a continuum from nearly aviru- tile degenerating cells when mature (Figure 15.2). Plaques
lent to maximally virulent. Pathotype classification are usually less than 1 mm in diameter and of variable
schemes have evolved over the last 30 years with the con- cell density, although plaque size varies with viral strain,
tinued increase in virulence. Current classification time, and other factors. Polykaryocytosis is seen in
schemes recognize four groups of viruses. These groups ­cultured fibroblasts and is a major component of the
are designated as mMDV, vMDV, vvMDV, and vv+MDV viral plaques or foci frequently used as a marker in virus
(597, 604). Pathotyping of virus isolates involves com- assays. Affected cells may contain two to several hun-
parative pathogenicity tests in vaccinated and unvacci- dred nuclei, and type A intranuclear inclusion bodies are
nated maternal antibody positive chickens with prototype commonly seen. Despite release of rounded cells into the
viruses as controls (592, 601). Detailed reviews on the medium as plaques mature, large areas of cell lysis are
increasing virulence and the details of prototype viruses not seen.
have been published (211, 498, 598, 599, 601). The evolu- Serotype 1 plaques develop in 5–14 days on primary
tion in the virulence of MDV strains is recognized, but isolation and in 3–7 days after adaptation to culture and
the molecular basis for this evolution has not been eluci- usually are enumerated by microscopic examination, but
dated, as genome sequence analyses of a number of these different staining techniques have been developed,
strains have not provided definite markers associated allowing enumeration at a later time. Differences in
with the virulence phenotypes. development and morphology of serotype 1 plaques in
Certain biologic characteristics are associated with chick and duck cells and in plaques induced by the three
pathotypes of serotype 1 MDVs but are most pronounced viral serotypes (484) have been described. Other cell cul-
between low‐passage and high‐passage (attenuated) ture systems such as chick embryo skin (445), tracheal
strains. Serial passage in vitro (30–70 passages usually explants (460), and embryo fibroblasts from several
are required) results in attenuation of virulent isolates avian species including Japanese quail (453) also have
(470, 494, 589). Attenuated strains grow more readily in been used.
vitro but produce lower viremia titers in vivo (609), A few avian cell lines such as OU2 (8, 9), quail muscle
which may be associated with a marked decrease in their cell line QM7 constitutively expressing MDV‐1gE (471,
ability to infect and/or replicate in lymphocytes and 508), JBJ‐1 (206), chicken embryo liver (327), and lines
spread between birds (160, 494, 591). There could also be derived from Muscovy duck retinal tissue (293) have also
incomplete attenuation resulting in minor lesions in sus- been used for the propagation of MDV strains with vary-
ceptible chickens (439, 575) or over‐attenuation result- ing success. Quail cell lines free of MDV can be used to
ing in low level in vivo replication and poor immune propagate serotype 1 MDV and HVT, but SB‐1 did not
response (318, 614). The in vivo growth potential of replicate efficiently (343). Recently developed keratino-
attenuated serotype 1 isolates can be increased by back- cyte cell lines support MDV replication, producing ­cell‐
passage in chickens (161, 591). A recent study on the associated viral progeny (140).
molecular basis of attenuation identified several de novo Serotype 1, but not serotype 2, MDVs can also be
mutations associated with reduced virulence (258). grown in chicken splenic lymphocytes in vitro (106).
Passages are made by the addition of fresh spleen cells to
the suspension cell cultures every two days, and infec-
Laboratory Host Systems
tion is monitored by IF. Herpesvirus of turkey may be
Marek’s disease virus usually is propagated and assayed similarly grown in turkey spleen cell cultures, but viral
in tissue cultures, newly hatched chicks, and embryo- antigen is rarely seen, if at all.
nated eggs. Lymphoblastoid cell lines from MD lympho-
mas are also an important laboratory host system. Chickens
Newly hatched chicks inoculated with virulent, serotype
Cell Cultures 1 MDV develop gross lesions or lesions that can be
The propagation of MDV serotypes in vitro has been detected histologically in ganglia, nerves, and certain
reviewed (484, 487, 498). Isolation of low‐passage MDV viscera after 2–4 weeks. Response is greatly dependent
in CEF or embryonal CKC cultures is far less efficient on genetic susceptibility of the chicken and virulence of
than in CKC or DEF (487) as propagation in CEF leads to the MDV isolate. Presence of virus or antibody, which
accelerated attenuation (494). In embryonal CKC, repli- can be detected by in vitro tests, or the presence of virus‐
cation of serotype 1 MDV (but not HVT) is abortive, associated antigen detected by FA tests on tissues, are
leading to loss of infectivity within two to three passages. also specific host responses of inoculated chickens to
Chapter 15  Neoplastic Diseases 559

(A) (B)

(C) (D)

(E) (F)

Figure 15.2  Focal lesions in cultured cells infected with various Marek’s disease virus (MDV) serotypes. (A) Low‐passage serotype 1 MDV
in chicken kidney cells cultured from an infected chicken, 9 days. (B) Low‐passage serotype 1 MDV in duck embryo fibroblasts (DEF), 5
days. (C) High‐passage, attenuated serotype 1 MDV in chick embryo fibroblasts (CEF), 5 days. (D) Low‐passage serotype 2 MDV in CEF, 8
days. (E) Low‐passage HVT (serotype 3) in CEF, 4 days. (F) Low‐passage turkey herpesviruses (HVT) in DEF, 12 days. All photos unstained,
about ×40. (Witter)
560 Section II  Viral Diseases

MD infection. All these responses are markedly enhanced


in chicks lacking maternal antibodies against MDV (86).
The induction of virus‐specific lesions in the wing web
(99) or the feather pulp (377) constitutes alternate
approaches that provide direct access to the site of lesion
development.

Embryos
Virus pocks develop on the chorioallantoic membrane
(CAM) of chicken embryos following yolk sac inocula-
tion with cellular MDV preparations (49, 574). Embryos
have also been used for MD vaccine evaluation, because
in ovo vaccination is becoming increasingly common in
the field. Embryos may also be used to isolate MDV
viruses that cannot be isolated directly in cell culture for
unknown reasons. Yamaguchi et  al. (633) reported the Figure 15.3  Smear from the MDCC‐RP1 cell line. Note the
isolation of MDV from the QT35 cell line by using kid- characteristic lymphoblastoid morphology and the mitotic
ney cell cultures prepared from 4‐ to 7‐day‐old chicks figures. Giemsa, ×1500. (Nazerian)
that had been inoculated at embryonic day (ED) 8 with
QT35 cells. gene expression from a number of MDV‐induced
tumors and transformed cell lines (358, 531, 559) will
Lymphoblastoid Cell Lines help to identify the molecular and biochemical path-
Lymphoblastoid cell lines developed from MD lympho- ways of transformation and the maintenance of the
mas grow continuously in cell culture without attach- transformed phenotype in these cells.
ment to the culture vessel. Success rates for establishing
cell lines from MD lymphomas have improved because
of better methodology (108, 428). Many cell lines are
now available including several from MD lymphomas in
Pathobiology and Epizootiology
turkeys (392). The majority of the chicken cell lines
Multiple Syndromes
established from lymphomas are CD4+/CD8‐ T cells
expressing major histocompatibility complex (MHC) Marek’s disease consists of several distinct pathologic
class II and T cell receptor (TCR) 2 or 3 (386, 417, 495). syndromes (90) with apparent differences among these
Lymphoblastoid cell lines can also be established from syndromes typically seen in commercial flocks,  and
lymphocytes harvested from early (4–6 days postinfec- those induced in the laboratory. Lymphoproliferative
tion) lesions induced in the wing web or pectoral muscle lesions, especially lymphomas, are most frequently
by injection of a mixture of MDV and allogeneic kidney associated with MD and have the most practical
cells. The cell lines from early lesions may be CD4+/ importance (Table  15.3A). However, skin leukosis
CD8−, CD4−/CD8+, or CD4−/CD8− (495). Cells of the in  broilers, fowl paralysis, persistent neurological
MDCC‐RP1 line are illustrated in Figure 15.3. disease, and ocular lesions are additional clinical
­
Some transformed cells contain about 5–15 copies of manifestations with lymphoproliferative components.
viral genome, although the mean number may be consid- Some of the lymphoproliferative syndromes may also
erably higher in different cell lines, perhaps in relation to have degenerative components. Several additional
the proportion of productively infected cells in the clinical syndromes characterized solely by degenera-
­population (376, 473). Most cell lines can be termed tive and inflammatory lesions, often with accompanying
“producer” lines, because a small proportion (1–2%) of immunosuppression, are induced by experimental
the cells enter into productive infection (108, 443). Viral infection (Table 15.3B). Non‐neoplastic brain pathol-
DNA can be highly methylated in cell lines although ogy, mainly vasogenic edema, is responsible for tran-
methylation is not essential for maintaining the trans- sient paralysis (231). Vascular lesions are manifested
formed state (298, 417). Methylation profiles of the viral as atherosclerosis (192). Under laboratory conditions,
genome in the cell lines are not very different from those young chicks inoculated with tumor cells may develop
in the primary tumors (63). transplantable tumors (267, 544, 558). Inoculation
Marek’s disease tumor-derived cell lines have been of  MDV‐infected, allogeneic CKC in the wing web
used to analyze the potential interaction with tumor may induce local tumor lesions (99). Some of the syn-
suppressor genes and cellular oncogenes, such as Meq. dromes induced under laboratory conditions are rare
The data generated from the systematic analysis of the or nonexistent in the field, probably because most
Chapter 15  Neoplastic Diseases 561

Table 15.3A  Clinical and pathologic syndromes associated with Marek’s disease virus (MDV) (part A).

Lymphoproliferative syndromesab (Marek’s disease)

Situation in which syndrome Lymphomas and nerve Fowl paralysis (nerve Skin leukosis Blindness and ocular
observed lesions lesions) (integument) lesions

Experimental chickens (laboratory)


Clinical signs Depression, death, Paralysis Swollen feather Blindness, ocular
stunting, paralysis follicles lesions
Mortality 0–100%c 0–30%cd None Rare or nonec
E
Age Onset 2–8 weeks PI Growing birds Young birds 4–8 weeks PI
Organ Visceral Mostly peripheral Skin Eye (iris, cornea)
organs+peripheral nerves
nerves
Layer/breeder flocks (field)
Clinical signs Depression, death, Paralysis, death Swollen feather Blindness, gray eye
paralysis follicles
Prevalence Common Occasionald Rare or nonee Rare
Mortality 0–60% 0–20% None None
Age 4–90 weeks 8–20 weeks 4–8 weeks PI >10 weeks
Broiler flocks (field)
Clinical signs Depression, death, Paralysis, death Swollen feather Blindness, gray eye
paralysis follicles, red leg
Prevalence Common Rare or noned Commone Rare or none
Mortality Minor — None None
Age At processing — At processing —
a
 Neoplastic lesions may include inflammatory components.
b
 Severity of syndrome usually less in vaccinated flocks.
c
 Depends on experimental conditions (virus strain, dose, chicken genotype, maternal antibody status, prior vaccination, etc.).
d
 Rarely induced by contemporary MDV strains, except in conjunction with visceral neoplastic lesions.
e
 Not usually recognized except at broiler processing or after feather removal.

commercial chickens are maternal antibody positive collected condemnation data from processing plants,
and vaccinated. including young broiler chickens condemned for leuko-
Subclinical syndromes may also occur but are more sis, which almost exclusively refers to MD. This data,
difficult to define. Vaccinated flocks produced more eggs analyzed and reported from the USDA National
than nonvaccinated flocks, indicating that MDV may Agricultural Statistics Service, shows a gradual decrease
depress productivity in otherwise normal‐appearing, in leukosis condemnations of young broiler chickens
nonvaccinated chickens (454). since the early 1970s, reaching an all‐time low of
0.00069% during 2016 (Figure 15.4). Regional differences
are striking, as illustrated by annual leukosis condemna-
Incidence and Distribution
tion rates in Delaware versus Georgia. The data has also
Marek’s disease exists in all poultry‐producing countries. revealed a biphasic pattern with maximum condemna-
Marek’s disease is generally considered ubiquitous tion rates around April and minimum frequencies
among vaccinated commercial flocks, although recent around August, although this seasonal trend has steadily
evidence suggests not all flocks are necessarily infected decreased since about 1990 (309, 498, 596).
(56, 581). Reporting systems vary, however, and it is dif- Sporadic outbreaks of MD occur on individual farms
ficult to determine the true MD incidence (174). Even in or regions. Recent industry‐wide surveillance study in
susceptible chickens, infection does not always induce Pennsylvania demonstrated the persistence of the virus
clinical disease and, in genetically resistant or vaccinated with fluctuation in virus loads at different farms (309).
chickens, infection may rarely cause overt disease. Several reports lend credence to the implication of
Since 1961, the United States Department of Agri­ exceptionally virulent MDV isolates in vaccine failures
culture (USDA) Food Safety and Inspection Service has (e.g., 35, 597), but it is important to exclude CIAV as a
562 Section II  Viral Diseases

Table 15.3B  Clinical and pathologic syndromes associated with Marek’s disease virus (MDV) (part B).

Lymphodegenerative Vascular
syndromes CNS syndromes syndromes Other syndromes

Early mortality syndrome, Transient paralysis and


Situation in which cytolytic infection, persistent neurological Local lesions;
syndrome observed immunodepression diseases Atherosclerosis transplants (transpl.)

Experimental chickens (laboratory)


Clinical signs Depression, stunting, Transient paralysis, tics, None Swelling at
death, increased disease torticollis, death inoculation site
susceptibility
Mortality 0–100%ab 0–100%ab None Yes (transpl.)
Age 9–20 days PI 9–28 days PI Adult birds Young birds
Organ Bursa, thymus, spleen Brain Blood vessels Web–local and
many–transpl.
Layer/breeder flocks (field)
Clinical signs Increased disease Transient paralysis, tics, — N/A: Only
susceptibility torticollis experimental
Prevalence Rarea Rarea Rare or none —
Mortality — Rare — —
Age — 5–12 weeks — —
Broiler flocks (field)
Clinical signs Increased disease Transient paralysis, tics, — N/A: Only
susceptibility torticollis experimental
Prevalence Rarea Occasionala None —
Mortality — Rare — —
Age — 5–7 weeks — —
a
 Not normally observed in chickens vaccinated for MD.
b
 Depends on experimental conditions (virus strain, dose, chicken genotype, maternal antibody status, prior vaccination, etc.).

10 Figure 15.4  Marek’s disease condemnations


in young broilers for the period 1961–2016
(data from National Agricultural Statistics
1 Service). (Dunn)

0.1
Percent

0.01

0.001

USA
0.0001
Delaware
Georgia

0.00001
1960 1970 1980 1990 2000 2010 2020
Chapter 15  Neoplastic Diseases 563

cofactor (370). It is usually difficult to associate increased types has provided measurements of virus load in feather
virulence of MDV isolates with regional fluctuations in pulp and dust (2, 31, 276, 469). These assays may measure
MD frequency, but layer outbreaks in Ohio in 1995 cell‐associated virus in the feather pulp and therefore do
yielded isolates of unusual virulence (95, 597). not necessarily provide a measure for the amount of infec-
In recent years, MD has been reported as the most tious virus shed into the environment. The latter question
commonly diagnosed infectious disease among backyard is relevant because dust collected from poultry houses
chickens in multiple countries (369, 438). containing HVT‐vaccinated flocks is positive for HVT
DNA. It is possible that this DNA is derived from cell‐
associated HVT orginally present in feather pulp cells
Natural and Experimental Hosts
rather than true cell‐free infectious virus. Older literature
Virtually all chickens including game fowl are susceptible had suggested that horizontal spread of HVT from vacci-
to MDV infection and tumor development (498) and are nated chickens is limited at best (498). Feather dust has
by far the most important natural host. Quail, turkeys, recently been used as a direct source for MDV whole
partridges, pheasants, and some species of ducks and genome sequencing for representing shed virus from a
geese are also susceptible to infection and disease (1, flock versus a point source in one animal (413).
499). Outbreaks in Japanese quail are most commonly Vertical transmission of MDV does not occur (470, 535,
reported among quail. In turkeys, occasional outbreaks 536). Transmission from dam to progeny as the result of
have recently been reported (54). Vaccination with external egg contamination is also unlikely because of
CVI988 appears to offer protection (137), whereas HVT, poor virus survival at temperature and humidity levels
interestingly, does not (182). Marek’s disease virus can be used during incubation (97). Passive transmission by dar-
re‐isolated from experimentally infected ducks (39), but kling beetles (Alphitobius diaperinus) has been reported,
development of MD has not been demonstrated. Most but free‐living litter mites, mosquitoes, and coccidial
other avian species including sparrows, pigeons, and pea- oocysts do not transmit MDV (40, 59, 60, 179).
fowl are probably refractory (reviewed in [499]). Recently Experimental transmission is commonly accomplished
a herpesvirus was isolated from three species of endan- by parenteral inoculation of susceptible chickens with
gered pheasants causing hepatocellular necrosis. Based cell‐associated virus from cell cultures, tumor cell sus-
on sequence analysis of two genes, this virus is related to pensions, or blood cells from infected chickens. Exposure
HVT and is proposed to be a new member of the genus by direct or indirect contact with infected chickens is
Mardivirus (510). Marek’s disease‐like lymphoid tumors also effective. Cell‐free but not cell‐associated virus can
in passerines are frequently cited, but need to exclude be used for intratracheal instillation or inhalation expo-
lymphoma‐like lesions caused by infection with Isospora, sure. An aerosol‐based exposure model was described
especially in captive populations (150). Mammals, includ- (6), which may facilitate studies on the early events in the
ing several species of primates, are refractory to experi- pathogenesis.
mental inoculation (reviewed in [499]).
Incubation Period
Transmission, Carriers, Vectors
The incubation period for experimentally induced MD is
Marek’s disease virus is transmitted readily by direct or well established (see reviews 27, 90). Mononuclear infil-
indirect contact between chickens by the airborne route trations containing AV37+CD4+ lymphocytes can be
(citations in 46, 499). The FFE produces fully infectious detected in nerves of maternal antibody‐negative, genet-
virus (92). These cells present in feathers and dander form ically susceptible chickens as early as 5 days PI (75).
the major source of contamination of the environment Monoclonal antibody AV37 recognizes chicken CD30hi,
and infection of chickens (41, 110). Contaminated poultry which is considered a marker for transformed lympho-
house dust remains infectious for at least several months cytes (78). Clinical signs and gross lesions generally do
at 20–25°C and for years at 4°C (citations in 89, 499). not appear until between the third and fourth weeks.
Under commercial conditions, young chickens are most The incubation periods can be short for several non-
commonly exposed to MDV by contact with r­ esidual dust lymphomatous syndromes associated with MDV infec-
and dander in the growing house or by aerosolized dust tion. Cytolytic infections occur at 3–6 days PI and are
from adjacent chicken houses, fomites, or personnel. After followed by degenerative lesions (atrophy) of the thymus
the virus is introduced into a chicken flock, regardless of and bursa of Fabricius within 6–8 days PI (74). The early
vaccination status or genetic resistance, infection spreads mortality syndrome (EMS) is characterized by deaths at
quickly from bird to bird. Early studies, based on contact 8–14 days PI (615). The clinical expression of both acute
infection, demonstrated that virus excretion begins about and classical forms of transient paralysis usually occurs
two weeks postinfection (310) and continued indefinitely from 8–18 days PI (311, 607) and can occur when SPF
(622). The development of qPCR assays for the three sero- birds are challenged with vv+MDV between 30 and 102
564 Section II  Viral Diseases

weeks of age (605). Field cases of transient paralysis are


seen mainly between 6–12 weeks of age, probably reflect-
ing MDV exposure 8–10 days prior to the onset of symp-
toms. Development of atherosclerosis requires 3–7
months (191). Induction of tumors within 10–14 days
after inoculation of cellular material is suggestive of a
transplantation response (544). Local lesions in the wing
web are visible 3–4 days PI with allogeneic MDV‐infected
CKC. Cell lines have been established from these lesions
suggesting that transformed cells are present (99).
Under field conditions, MD outbreaks sometimes
occur in unvaccinated layer chickens as young as 3–4
weeks. Most of the serious cases begin after 8–9 weeks
but sometimes commence well after the onset of egg Figure 15.5  Fowl paralysis. Spastic paralysis of limbs associated
production, especially in broiler breeders (381) or subse- with peripheral nerve involvement in Marek’s disease. (Witter)
quent to molting (600). Witter (600) differentiated the
outbreaks in commercial, vaccinated chickens as “early”
or “late” breaks. Witter and Gimeno (605) suggested that the central nervous system (CNS) signs are difficult to
late breaks were not likely the result of recent infections distinguish from those associated with MD nerve
alone and that additional factors are needed to cause the lesions.
late breaks. The different manifestations of MD, includ- Birds with ocular involvement may show evidence of
ing EMS (113), may occur in backyard flocks, which are blindness (198, 415, 543). Ficken et al.(198) isolated two
often not vaccinated. MD strains from commercial flocks with greater than
90% blindness. The blindness was reproduced in experi-
mentally‐infected commercial chickens. Gross ocular
Clinical Signs
lesions were not always present in blind birds. The blind-
Signs associated with MD vary according to the specific ness can be unilateral or bilateral, although recognition
syndrome (Tables 15.3A and 15.3B). Chickens with MD of clinical blindness requires careful observation.
lymphoma or fowl paralysis syndromes may exhibit Affected eyes gradually lose their ability to accommo-
signs, but few are specific to MD (45). In general, signs date to light intensity.
related to peripheral nerve dysfunction are those asso- Early mortality syndrome results in high mortality
ciated with asymmetric progressive paresis and, later, 8–16 days PI of young chickens with virulent MDV strains
complete spastic paralysis of one or more of the extrem- (571, 615). Chickens become depressed and comatose
ities. Involvement of the vagus nerve can result in prior to death, which occurs within 48 hours of the onset
paralysis and dilation of the crop and/or gasping. of signs. Some affected chickens may also exhibit flaccid
Because locomotory disturbances are easily recognized, neck paralysis prior to death (607). Chickens undergoing
incoordination or stilted gait may be the first observed acute cytolytic infection at 3–6 days PI may be depressed
sign. A particularly characteristic clinical presentation but rarely die during this period, although some may die
is a bird with one leg stretched forward and the other later from EMS. Immunosuppressed chickens may suc-
back as a result of unilateral paresis or paralysis of the cumb to ancillary infections, but some chickens die 20–40
leg (Figure 15.5). However, chickens with MD lympho- days PI with few signs.
mas may appear clinically normal but have extensive Classical and acute transient paralysis syndromes
neoplastic involvement when euthanized, while other have been described in field flocks (643) and is associ-
birds may become depressed and comatose prior to ated with MDV infection (311). It has been observed
death. Nonspecific signs such as weight loss, paleness, infrequently in the field since vaccination for MD has
anorexia, and diarrhea may be observed, especially in become widespread, but may be encountered in non‐
birds in which the course is prolonged. Under commer- vaccinated backyard flocks. In the classical form,
cial conditions, death often results from starvation and affected chickens display varying degrees of ataxia and
dehydration because of the inability to reach food and flaccid paralysis of the neck or limbs beginning 8–12
water or from trampling by flockmates. Some birds days PI (Figure 15.6). Signs typically last 1–2 days fol-
develop nervous tics or torticollis 18–26 days PI, often lowed by a rapid and complete recovery, although
after recovery from classical transient paralysis. This recovered chickens may succumb a few weeks later
syndrome, termed persistent neurological disease with MD lymphomas. The acute (fatal) form results in
(231), can be induced by partially attenuated MDVs death within 24–72 hours following the onset of para-
that no longer induce ­transient paralysis (229). However, lytic signs (607).
Chapter 15  Neoplastic Diseases 565

vaccination (408, 604). The extent of disease induced by


a given strain depends in part on the genetic constitution
of the host (493).

Virus Dose and  Route.  Dosage may influence disease


frequency under natural conditions, although the MD
response in genetically susceptible birds given virulent
virus was found to be maximal even when a limiting
dilution of virus was inoculated (532). Infection with
cell‐free MDV through intratracheal inoculation or by
aerosol may enhance early virus replication and increase
the development of lymphomas compared to parenteral
Figure 15.6  Transient paralysis. Flaccid paralysis of neck of young inoculation (6, 19, 84).
chicken nine days after inoculation with Marek’s disease virus.
(Courtesy of Avian Diseases.) (Witter)
Host Gender.  Several studies indicated that females died
earlier and experienced higher losses than males (357,
Morbidity and Mortality 427), but the opposite has also been reported (363). The
The incidence of MD is quite variable in commercial differences were apparently not due to sex hormones,
flocks and in general low since the worldwide introduc- varied with the genetic strain, and were most pronounced
tion of vaccines, although problems are sometimes with genetically susceptible chickens and with viruses of
reported (174, 381). Most birds developing clinical dis- higher virulence. In practice, the influence of gender is
ease die. A few birds may apparently recover from the probably less important.
clinical disease (52, 75), but the recovery is rarely perma-
nent. Prior to the use of vaccines, losses in affected flocks Maternal Antibodies.  Maternal antibodies reduce and
were estimated to range from a few birds to 30% and delay MD mortality (129), EMS (615), and transient
occasionally as high as 60% (381). In broilers, MD con- paralysis (311), probably by limiting, but not preventing,
demnations averaged 1.0% in 1970 with 10% or higher in the spread of virus in tissues during the first few days
individual flocks (452). The average condemnation rate post  exposure (86, 429). Thus maternal antibodies do
for MD in the United States has decreased dramatically not  provide a sterilizing immunity. Breeder stocks are
(see Incidence and Distribution). vaccinated uniformly and exposed to virulent MDV and
Some flocks experience significant disease outbreaks virtually all chickens are hatched with maternal antibodies
despite vaccination. After the disease appears, mortality against multiple serotypes. Specific pathogen free flocks
builds gradually and generally persists for 4–10 weeks. are a source of antibody‐free chicks for laboratory studies.
Outbreaks occur and abate spontaneously in isolated
flocks or occasionally in several flocks in a region or in Host Genetics and  Age at Exposure.  Genetic factors (see
succeeding flocks on a farm. The reasons for these fluc- reviews [20, 71]) and age at initial exposure are important
tuations are poorly understood. determinants of MD susceptibility (see Chapter 2).
Response rates or mortality approaching 100% for Age‐related resistance is an expression of genetic
lymphomas, EMS, acute cytolytic infection, or transient resistance and develops after hatching paralleling the
paralysis can be achieved following inoculation or expo- development of immune competence. Newly hatched
sure of unvaccinated, susceptible chickens to MDV. chicks and older chickens are both susceptible to infec-
Because the response frequency is influenced by many tion and cytolytic infection (90), but cytolytic infections
factors (see Factors that Influence Mortality and Lesions), are resolved more rapidly in older birds (82) and virus
laboratory experiments can be designed to produce a load is somewhat lower (156). The frequency of lympho-
wide range of specific clinical and pathologic responses. mas is variable and often markedly reduced in older
chickens compared to newly hatched chicks, especially
Factors that Influence Mortality and Lesions in genetically resistant lines (498). However, non‐vacci-
Virus Strain.  The virulence of MDV strains varies widely nated, SPF, older chickens may develop high rates of lym-
and appears to have increased over time (597) and is phomas and transient paralysis following challenge with
reviewed in (159, 489). Compared to the milder forms of vv and vv+ strains (269, 605). Age‐related resistance can
the disease, which caused mainly peripheral nerve be abrogated by neonatal thymectomy (520) suggesting
lesions, very virulent (vv) and vv+ pathotypes frequently that other immunosuppressive factors, for example,
induce higher mortality and more visceral lymphomas, CIAV (501), may increase the susceptibility of older
and have the tendency to more frequently break through chickens to disease. Lesion regression has been linked to
genetic host resistance or immunity induced by age‐related resistance (75, 519).
566 Section II  Viral Diseases

Prior Infection.  Early studies had shown that mild,


serotype 1 strains can induce protective immune
responses against challenge (533). Dunn et  al. (176)
demonstrated superinfection following a short interval,
but if the second challenge was given after 14 days,
regardless of virulence, superinfection was significantly
reduced.

Environmental Factors and Stress.  Various environmental


factors and intercurrent infections appear to affect
the  incidence of MD, probably through interference
with immune responses. Gross (235) observed increased
incidence among chickens selected for high concentra­
tions of plasma corticosterone or subjected to a high
degree of social stress. The administration of cor­
ticosteroids to latently infected chickens precipitated the
appearance of clinical MD (441). Feeding of corticosteroid Figure 15.7  Enlarged sciatic plexus (left) and normal plexus
(right). (Peckham).
inhibitors tended to increase resistance to MD (134).
Restricted feed intake delayed and reduced incidence
of  MD (240) whereas high‐protein diets (450) or the controls to detect changes. Careful examination of the
selection for fast growth rate were associated with various nerve ramifications may be necessary to expose
increased susceptibility to MD (240). gross lesions in some birds, because enlargements can
Because MDV infection may depress host immune vary in both presence and degree from one portion of an
responses in its own right (see Immunosuppression), affected nerve to another. Figure 15.7 illustrates unilateral
concurrent infections are often exacerbated. However, gross enlargements in the sciatic plexus.
when the concurrent infection is itself immunosuppres-
sive, the resulting immunosuppression usually will exac- Visceral Organs.  Lymphomas may occur in one or more
erbate both disease processes. Examples include IBDV, of a variety of organs and tissues. Lymphomatous lesions
REV, and CIAV (291, 578, 610, 648). Problems with CIAV can be found in the gonad (especially the ovary), lung,
contamination invariably interfere with the evaluation of heart, mesentery, kidney, liver, spleen, bursa, thymus,
MDV stocks for relative virulence (371). adrenal gland, pancreas, proventriculus, intestine, iris,
skeletal muscle, and skin. Probably no tissue or organ is
without occasional involvement. Both the genetic strain
Pathology
of chicken and the virus strain can influence the organ
Gross Pathology distribution of lesions. Visceral lymphomas are common
Pathologic changes in MD have been reviewed (425, 427) in more virulent forms of the disease (597). Visceral
and consist mainly of nerve lesions and visceral lympho- tumors can occur in the absence of gross nerve lesions,
mas. Macroscopic changes are not seen in the brain, but especially in certain strains of chickens. Marek’s disease
gross enlargements can be found in spinal ganglia. lymphomas in most viscera appear as diffuse
enlargements, sometimes to several times the normal
Nerves.  Severely affected peripheral nerves may show size, and a diffuse white or grayish discoloration is often
loss of cross‐striations, gray or yellow discoloration, and present (Figure  15.8B). Alternatively, lymphomas may
sometimes an edematous appearance. Usually, plexi of occur as focal, nodular growths of varying size
the sciatic and brachial nerves are more enlarged than (Figures 15.8E and 15.8F). Nodules are white or gray in
the respective trunks. Localized or diffuse enlargement color and are firm, and the cut surface is smooth.
causes the affected portion to be 2–3 times normal size, Necrosis is rare but may occur in the center of rapidly
in some cases much more. Goodchild (233) reported growing lesions.
that autonomic nerves and especially the celiac plexus Diffuse infiltration of the liver causes loss of normal
are affected at a higher frequency than peripheral nerves, lobule architecture and often gives the surface a coarse
for example, the sciatic and brachial nerves. Witter (595) granular appearance. Nodular tumors may also be seen
found the cervical vagus to be of particular diagnostic in the liver. Lesions in the immature ovary are observed
importance. Because unilateral and or minimal as small to large grayish translucent areas (Figure 15.8B).
enlargements may be important indicators of disease, it With large tumors, the normal foliated appearance of the
is helpful to examine opposite nerves and, in experimental ovary is obliterated. Mature ovaries may retain function,
infections, to compare with age‐matched normal even though some follicles are tumorous. Marked
Chapter 15  Neoplastic Diseases 567

involvement is indicated by a cauliflower‐like appear- response to viral replication and is induced by all three
ance. The proventriculus becomes thickened and firm as serotypes. Vascular syndromes are manifested principally
a result of focal leukotic areas within and between the by occlusive atherosclerosis (192). Susceptible P‐line
glands, which may be seen through the serosal surface or, chickens inoculated with the CU2 isolate of MDV
if involvement is diffuse, detected by palpation. Affected developed grossly visible fatty atheromatous lesions in
hearts are pale from diffuse infiltration or have single large coronary arteries, aortas, major aortic branches, and
or  multiple nodular tumors in the myocardium other arteries (Figure 15.8D). Lymphoid tumor transplants
(Figure 15.8F). Pinpoint foci may be seen in the epicar- and local lesions are experimental syndromes characterized
dium. Involvement of the lung (Figure  15.8E) may be by nodular growths at the site of inoculation, although
indicated by increased firmness of the organ upon palpa- some transplantable tumors metastasize readily to the
tion. Muscle lesions may be present in both superficial liver and spleen, causing diffuse enlargements (544).
and deep layers and are most common in the pectoral
muscle (43). Gross changes vary from tiny whitish streaks Microscopic Pathology
to nodular tumors. Histopathologic changes associated with MD lym-
phoproliferative lesions have been described by numer-
Integument.  Skin lesions, probably the most important ous workers who are in general agreement about the
cause of condemnation in broiler chickens, usually are types of histologic lesions and the cells involved (reviewed
associated with feather follicles. The nodular lesions may in 424, 425, 427, 499).
involve few scattered follicles, or they may be numerous
and coalesce. The distinct whitish nodules (Figure 15.8A), Nerves.  In peripheral nerves, two main types of
especially evident in dressed carcasses, may become lymphoproliferative lesions are recognized, which are
scablike with brownish crust formation in extreme cases referred to as type A and B, respectively. Type A lesions
(43). Lapen and Kenzy (325) found the highest incidences consist mostly of CD30+CD4+ T cells (75) and some B
of lesions in external and internal crural and dorsal cells and is considered neoplastic. In some cases,
cervical tracts. Erythematous involvement of the shank demyelination and Schwann cell proliferation are
integument is seen, especially in virulent forms of the associated with A‐type lesions. The second, B‐type,
disease in broiler chickens (177) and is commonly known lesions are essentially inflammatory and are characterized
as “Alabama redleg.” Swelling of the comb or wattles may by diffuse, light‐to‐moderate infiltration by small
indicate lymphoma growth in underlying tissues (177, lymphocytes and plasma cells, usually with edema, and
180). Interestingly, FFE is not essential for virus sometimes, with demyelination and Schwann cell
production or development of skin tumors, because proliferation. A few macrophages may be found. The two
scaleless chickens produced cell‐free infectious virus in types may be observed in different nerves of the same
epithelial cells and developed skin tumors (250). bird or even in different areas of the same nerve. Lawn
and Payne (326) observed cellular infiltrations as early as
Eye.  Gross ocular changes, including loss of five days PI, which gradually increased in intensity until
pigmentation in the iris (“gray eye”) and irregularity of three weeks when severe proliferative, type‐A lesions
the pupil, are caused by mononuclear infiltration of the were seen in the absence of paralysis or demyelination.
iris (Figure  15.8C). Nearly all field isolates can induce Coincident with initial neurologic signs seen at four
ocular lesions in nonvaccinated or HVT‐vaccinated weeks PI, areas of widespread demyelination could
chickens (597). Conjunctivitis, occasionally with be  found within the proliferative lesions. Finally,
multifocal hemorrhages and corneal edema have also characteristic inflammatory, type‐B lesions (edema,
been observed (198). sparse infiltrations) appeared, suggesting the occurrence
of regression of A type lesions. A third, C‐type, lesion
Other Syndromes.  Gross lesions are associated with at consists of light infiltration of lymphocytes and plasma
least some of the other MDV‐associated syndromes. The cells. The sequence of events has been reviewed in detail
lymphodegenerative syndromes, related to intense by Payne et al. (427). Characteristic changes in nerves are
cytolytic infections of lymphoid organs, usually are illustrated in Figure 15.9.
characterized by severe bursal and thymus atrophy. The
cytolytic infection is first evident 3–6 days PI, but may Brain.  The principal CNS lesion in MD consists of mild,
persist becoming more obvious at 8–14 days PI (95, 571, but persistent, perivascular cuffing usually accompanied
615). After inoculation with highly virulent field strains, by gliosis but without primary demyelination as the
some chickens may die at 20–50 days without gross lesions principal CNS lesion in MD (231, 427). Experimental
except severe bursal and thymic atrophy (597). Some infection with less virulent MDV strains only resulted in
chickens also develop a transient splenomegaly within transient, moderately severe inflammatory lesions as
4–12 days PI (93). The splenomegaly is a non‐neoplastic early as 7–10 days PI. In contrast, lesions induced by
(B)

(A)

(C) (D)

(E) (F)

Figure 15.8  (A) Leukotic tumors involving feather follicles (skin leukosis). (Peckham) (B) Experimentally induced Marek’s disease (MD)
lymphoma in immature ovary (bottom) compared with normal ovary (top) (Witter). (C) Ocular lesions of MD. Note that the normal eye
(left) has a sharply defined pupil and well‐pigmented iris. Affected eye (right) has a discolored iris and very irregular pupil as a result of
mononuclear cell infiltration. (Peckham). (D) Gizzard from a chicken infected with CU‐2 isolate of MDV. Note the grossly obvious
atherosclerotic change in the arteries. (C. Fabricant). Microscopic changes from similar arteries are shown in Figure 15.16B. (E) Multiple
lymphomas in lungs. (F) Multiple lymphomas in heart. (Shivaprasad) (For color detail, please see the color section.)
Chapter 15  Neoplastic Diseases 569

(A) (B)

Figure 15.9  Microscopic lesions of Marek’s disease in peripheral nerves. (A) Type A lesion characterized by marked cellular infiltration,
numerous proliferating lymphoblastic cells, and no edema. H&E, ×550. (B) Type B lesion with edema, scattered infiltrating small and
medium lymphocytes, and plasma cells. H&E, ×420. (Gimeno)

Figure 15.10  Extensive infiltration of lymphoblasts extending


into the neuropil in the cerebellum of a Marek’s disease virus‐
infected chicken three weeks postinfection (PI). H&E, ×400.
(Gimeno)

vv+MDV strains appeared earlier, were more extensive,


and were followed by proliferative lesions (227). The
initial lesions involve vascular elements; endotheliosis
occurs at 6 days PI and is followed at 8–10 days PI by a
moderate to severe infiltration of lymphocytes and
macrophages around blood vessels and scattered
throughout the neuropil (227). The vasculitis and edema Figure 15.11  Lymphoid cell infiltration of ovary. Organ is
composed largely of tumor cells, but a few ovarian follicles can be
disappear and may be followed by lymphoproliferative seen. H&E, ×116. (Witter)
infiltrations of large lymphocytes and glial cells. These
lesions tend to persist and are associated with persistent
neurological disease. Thus, as in nerves, brain lesions are Visceral Organs.  Lymphomatous lesions in visceral
both inflammatory and lymphoproliferative. Severe organs (Figures  15.11 and 15.12) are more uniformly
lymphoid infiltration in the cerebellum is shown in proliferative in nature than those in nerves and similar
Figure 15.10. in appearance to A‐type lesions consisting of diffusely
570 Section II  Viral Diseases

and infiltration of mostly CD8+ T cells, plasma cells,


heterophils, and macrophages. The cellular infiltration
probably represented an immune response to MDV viral
antigens such as pp38, which is recognized by CTL (404).
The late lesions appeared between 26–56 days PI and
consisted of uveitis, keratitis, and retinal necrosis.
Infiltrating cells included CD4+ and CD8+ T cells,
macrophages, plasma cells, and granulocytes. Early and
late lesions did not contain transformed lymphocytes
based on the absence of meq expression.
The unusually severe ocular lesions described by Ficken
et al. (198) include uveal changes with increased aqueous
humor protein, vascular engorgement, mild hyperemia to
severe swelling of the iris, severe inflammatory changes,
Figure 15.12  Higher magnification of a kidney lymphoma and edema of the cornea, including intranuclear ­inclusion
showing pleomorphic tumor cells. Kidney tubules (bottom) show
degeneration caused by tumor cell pressure. H&E, ×450 (Gimeno) bodies.

Blood.  Leukemia consisting of T lymphoblasts may


proliferating small to medium T lymphocytes and occur occasionally (100, 430). Extravascular hemolytic
lymphoblasts, NK cells, B cells and macrophages, while anemia has been reported after infection with vvMDV
plasma cells are rarely present (reviewed in 73, 427). strains in the absence of CIAV (209). The importance of
The cellular composition of tumors is similar from one this finding is not clear because hematocrit values are
organ to another, even though the gross pattern of not routinely used as a MD parameter. Jakowski et  al.
involvement may vary. Tumor cells are characterized by (283) reported MDV caused anemia as a consequence of
large, pleomorphic nuclei with prominent nucleoli bone marrow aplasia, but their MDV was later found to
(169). The majority of transformed T cells express be contaminated with CIAV (501). Blood lymphocyte
MHC class II and CD4 although CD4−CD8+, CD4+CD8+, numbers may vary during the infection cycle: T cells may
and CD4−CD8− T cells can also be transformed by be elevated while B cells are decreased (430). However,
MDV (495). In addition the tumor cells express high Morimura et  al. (378) reported an initial increase of
levels of CD30 (77) and resemble Treg cells (511). CD4+TCRαβ1 cells around 16 days PI followed by a
Pradhan et  al. (444) found immune complexes in the decrease at 30 days PI. Infection with vv+ C12/130
kidney, leading to glomerulopathy, in MDV‐infected caused significant increases in the absolute number of
chickens. They suggested that these lesions might be blood monocytes around 8 days PI. B cells, CD4+ and
one of the major causes of death in MD. CD8+ T cells decreased during the early cytolytic
infection followed by an increase between 8–10 days PI,
Integument.  Lymphoproliferative nodules often sur­ but these changes were also seen for T cells after infection
round feather follicles that contain MDV viral antigens with HPRS‐16 (37).
and intranuclear inclusions in the FFE (92, 127). These
lesions in the skin appear largely inflammatory but may Lymphodegenerative Syndromes.  Marek’s disease virus
also be lymphomatous. Massive proliferative lesions may replication in the bursa of Fabricius and thymus results
cause disruption of the epidermis, resulting in an ulcer. in transient acute cytolytic changes accompanied by
Moriguchi et al. (377) described both inflammatory and atrophy (citations in 89, 427). In experimental infections,
lymphoproliferative lesions in the feather pulp; the latter bursal lesions consist of follicular degeneration,
were closely related to the incidence of MD and may be lymphoid necrosis with depletion, and cyst formation
useful for antemortem diagnosis (128). (Figure  15.13A). Thymic atrophy is often severe, and
lymphocytes are depleted in both cortex and medulla
Eye.  Smith et al. (534) and Pandiri et al. (414) examined (Figure 15.13B). Intranuclear inclusions can sometimes
the histology of eye lesions after experimental infection be found in cells associated with degenerative lesions.
with the GA and Md5 strains, respectively. The former Viral antigens such as pp38 can be abundant during the
group found that apparently transformed cells could be acute cytolytic phase in infections with vv+MDV strains,
detected as early as 11 days PI in the arachnoid layer of especially in the medullary regions of the thymus and
the optic nerve and subsequently in ciliary nerves and bursal follicles (Figures 15.14A and 15.14B). Infection in
uvea. Pandiri et  al. (414) distinguished early and late the absence of maternal antibodies may cause focal or
lesions. The early lesions, between 6–11 days PI, generalized necrosis in a variety of organs, including the
consisted of hypertrophy of endothelial cells, vasculitis, kidney (86, 199). Following the acute cytolytic phase,
Chapter 15  Neoplastic Diseases 571

(A) (B)

Figure 15.13  Degenerative lesions in bursa and thymus of chickens inoculated with the 648A (vv1) strain of Marek’s disease virus.
(A) Bursa at 10 days postinfection (PI) shows degeneration and atrophy of follicles. (B) Thymus at 6 days PI shows necrosis and lymphoid
cell depletion. ×12 (Gimeno)

(A) (B)

Figure 15.14  Acute cytolytic infection of lymphoid tissues 6 days postinfection (PI) with the 648A (vv+) strain of Marek’s disease virus. The
pp38 viral antigen is visualized by immunohistochemical staining (black). (A) Bursa of Fabricius. (B) Thymus. ×30 (649). (Witter)

antigen positive cells disappear, and at least partial (549). Other, apparently unrelated, brain lesions
repopulation with lymphocytes occurs. Bursal and (perivascular cuffing, lymphocytosis, and gliosis) may be
thymic atrophy, however, may persist for several weeks observed after clinical recovery or in infected but
or longer. In the bursa, some interfollicular lymphoid clinically normal birds.
infiltration with T cells may occur. Early mortality
syndrome is characterized by severe lymphoid Vascular Syndromes.  Arterial lesions associated with
degeneration and death, often with enlarged, necrotic MDV‐induced atherosclerosis include proliferative and
spleens (615). Early mortality syndrome has been linked fatty‐proliferative changes in aortic, coronary, celiac,
with CNS signs and transient paralysis (342, 607). gastric, and mesenteric arteries (192, 372) (Figures 15.16A
and 15.16B) (557). Internal and medial foam cells,
CNS Syndromes (Transient Paralysis).  Lesions in classical extracellular lipid, cholesterol clefts, and calcium
and acute transient paralysis are very similar. The critical deposits characterized the fatty‐proliferative lesions.
lesion in both types is vasculitis (Figure 15.15) leading to Marek’s disease virus antigens could be detected by IF
vasogenic brain edema (231, 550, 607). Leakage of adjacent to the arterial lesions.
albumin and IgG around affected vessels results in
vacuolization. Edema and vasculitis develop coordinately Tumor Transplants and  Local Lesions.  Tumor transplants
with clinical flaccid paralysis and resolve in 2–3 days are composed of uniform, lymphoblastic cells with few, if any,
572 Section II  Viral Diseases

infiltrating host cells. In regressing tumor transplants small Pathogenesis


lymphocytes, heterophils, vascular invasion, and necrosis
Several reviews on the pathogenesis of MD have been
may be present (200). Local lesions induced in the wing
published (27, 90, 502), which also provide references for
web or pectoral muscle by inoculation of MDV‐infected,
older reviews. The use of BACs and overlapping cosmid
allogeneic CKC are inflammatory in nature, consisting
technologies has allowed the deletion of specific genes.
of  lymphocytes and macrophages and  sometimes
Using these technologies, the importance of several
accompanied by hemorrhages and necrosis (99).
genes for the pathogenesis has been established.
Four phases of infection in vivo can be delineated. (1)
Early productive–restrictive virus infection causing
primarily degenerative changes. The infection is
­
­productive–­restrictive because the virus remains cell‐
associated and is only transferred by cell to cell contact.
(2) Latent infection. (3) A second productive–restrictive
infection phase coincident with permanent immunosup-
pression. (4) The proliferative phase involving nonpro-
ductively infected lymphoid cells that may or may not
progress to the point of lymphoma formation
(Figure 15.17). This Division is somewhat arbitrary and
phases 2–4 can coexist in different cells in the same bird.
Infection with some of the vv+ strains may not follow
this general pattern, and mortality can occur without
even entering into the latent phase. The next section will
describe the classical pathogenesis in lymphoid tissues,
Figure 15.15  Transient paralysis lesions in the brain. Vasculitis in based mostly on studies in SPF chickens. The pathogen-
the cerebellum at 10 days postinfection (PI) showing endothelial
cell necrosis, lymphoid cell accumulations, and vacuolization.
esis of infection in the FFE involves epithelial cells and
Note intramural necrotic debris (arrow) as well as infiltration of will be discussed in the section on cytolytic infection
heterophils in the vessel wall. H&E, ×250. (Gimeno) in FFE.

(A) (B)

Figure 15.16  (A) Gastric artery of normal chicken. (B) Atherosclerotic artery in gizzard of chicken infected with CU2 isolate of Marek’s
disease virus. Lumen is occluded by thickened intima, and atheromatous changes have occurred deep in the intima and media. H&E, ×24.
(C. Fabricant)
Chapter 15  Neoplastic Diseases 573

Infection by inhalation
of poultry dust

Virus shedding Early replication


from feather in lungs
follicle epithelium
Environment

Infection

MD lymphoma
B-cell
Latent phase: B
lymohocytes Early Cytolytic phase
“cell death”

infiltration T
of nerves Activated T-cell
Transformation T
of lymphocytes “cell death”
Latently-infected
(3/4 weeks) T-cell 1 week

Figure 15.17  Schematic diagram showing the different stages of Marek’s disease (MD) pathogenesis including the virus shedding from
the feather follicle epithelium and the transformation of T lymphocytes in susceptible birds.

Early Productive‐Restrictive Infection (Phase 1) centage of CD4+ and CD8+ T cells expressing TCRαβ can
The virus enters the host via the respiratory tract and become cytolytically infected during the early phase of
cell‐free virus reaches the lymphoid organs within 24–36 the pathogenesis. The consequence is a transient atrophy
hours after intratracheal inoculation (6, 482). Marek’s of the lymphoid organs, especially the thymus and the
disease virus is probably transferred to the lymphoid bursa. Depending on the virulence of the challenge
organs by phagocytic cells (90). Based on the presence of strain, birds may recover between 8–14 days PI, or the
MDV proteins (36, 225) and transcripts (19) mac- atrophy may become permanent (93, 95). The cytolysis is
rophages can become infected, but because virus parti- likely initiated by the activation of the host shut‐off pro-
cles could not be demonstrated, it is not sure if this is an tein leading to cell‐death by apoptosis (379). Although
abortive or productive–restrictive infection. MDV‐infected cells in the thymus are mostly B cells
Shortly after infection either by inhalation of cell‐free (521), thymocytes undergo massive apoptosis possibly as
virus or by parenteral inoculation with cell‐associated the consequence of viral infection (29) or virus‐induced
virus, cytolytic infection can be detected in the spleen, cytokine changes.
bursa of Fabricius, and thymus, peaking between 3–6 During phase one hyperplasia of lymphoid and reticu-
days. Splenectomy (483) and embryonal bursectomy lum cells occurs (427), causing splenomegaly between
(EBx) (491) delayed or reduced productive–restrictive 4–7 days PI, which is also observed after infection with
infection and the development of lymphomas suggesting MDV‐2 and HVT (93). The level of infection is in general
a central role for these two organs in the pathogenesis of similar in genetically resistant and susceptible strains
MD. In contrast, neonatal thymectomy enhanced the during phase 1 (7, 295, 642). However, genetically resist-
pathogenicity of the low‐oncogenic CU‐1 and CU‐2 ant line 6 chickens have a significantly lower level of
strains (94). The explanation for these findings was pro- infected lymphocytes than susceptible line 7 chickens.
vided by Shek et  al. (521) discovering that the primary Interestingly, line 6 has more B cells than line 7, thus the
target cells in all three organs are B cells. Recently it has difference is not caused by a lack of target cells in line 6
been shown that B cells are also infected in the lung two (29). In spleens of line 7 birds, dramatic changes occur
days after intratracheal inoculation (19). Infection of T with irregular patches of pp38+ B cells becoming sur-
cells requires activation based on the expression of MHC rounded by TCRαβ1+ CD4+ and CD8+ cells, thus provid-
class II antigens and activated but not resting T cells can ing optimal conditions for virus transfer from B to T cells.
undergo cytolytic infection (102, 104, 105). Baigent and These data suggest that MDV may replicate and spread
Davison (29) confirmed that the early cytolytic infection more efficiently in line 7 than line 6 chickens (29, 104).
occurs mostly in B cells using dual staining techniques Recently several studies have been conducted to ana-
with mAb specific for B‐ and T‐cell markers and MDV lyze the changes in transcriptome and proteome in
pp38. In addition, they demonstrated that a small per- ­various tissues after MDV infection (152, 242). Changes
574 Section II  Viral Diseases

have been found for many genes and pathways, but addi- The extent to which nonlymphoid cells are latently
tional studies will be needed before general conclusions infected is not known, although apparent latent infection
can be drawn for the understanding of the pathogenesis has been observed in Schwann cells and satellite cells in
and immune responses. spinal ganglia (434).
Several factors can modify the early pathogenesis.
Prior vaccination or the presence of maternal antibodies Second Phase of Cytolytic Infection (Phase 3)
reduce the cytolytic infection (90). The reduction in The second cytolytic infection phase has not been stud-
cytolytic infection also will reduce the number of latently ied in great detail and does not always occur depending
infected cells and reduce or delay tumor development. on genetic resistance of the host and the virulence of
Exposure at one day of age prolongs the cytolytic infec- MDV. If present, localized foci can be found in the lym-
tion compared to exposure at two or seven weeks of age phoid organs and in tissues of epithelial origin in various
(82). Likewise, the pathogenicity of the virus strain may visceral organs (e.g., kidney, pancreas, adrenal gland, and
affect the severity of early infection. The vv and vv+ proventriculus). Focal cell death and inflammatory reac-
strains can cause more severe lymphoid organ atrophy tions develop around affected areas (10, 90). If the sec-
than the less oncogenic strains, resulting in an early mor- ond cytolytic infection occurs, many of the cytokines
tality syndrome (95, 615). upregulated during the first cytolytic phase are again
The apoptosis of lymphocytes during the early cytol- upregulated (422).
ytic phase (379) may cause transient or permanent
immunosuppression, depending on the virulence of the Cytolytic Infection in FFE
challenge strain. In addition, a transient suppression of Starting around 14 days PI cytolytic infection occurs in
mitogen stimulation has been reported, but this may the FFE (96), which is the only known site of complete
actually represent a protective response (497, 502). virus replication. The replication occurs in genetically
resistant as well as susceptible birds independently of the
Latent Infection (Phase 2) virulence of the MDV strain. Marek’s disease virus most
At about 6–7 days, the infection becomes latent when likely is transferred to the FFE by infected lymphocytes.
cytolytic infection can no longer be demonstrated, and Viral DNA of all three serotypes can be detected in
tumors are not yet detectable. The development of feather tips as early as 5–7 days PI by qPCR (see
latency coincides with the development of immune Transmission, Carriers, Vectors). It is not known if this
responses. The integration of the virus into the host tel- really represents infectious cell‐free virus or viral DNA
omeres is considered a key virus–host interaction for inside feather tips, because MDV‐positive peripheral
achievement of latency (367). Impairment of cell‐medi- blood leukocytes (PBL) can be found in feather pulp as
ated immunity (CMI) (82) or infection with the more early as 4 days PI concomitant with increased IFN‐γ
virulent pathotypes delays the onset of latency (642). transcription (4).
Several soluble factors have been implicated in the Lymphocyte aggregates consisting of small lympho-
induction of latency including IFN‐α, IFN‐γ, latency cytes with nuclear inclusions can be detected in the peri-
maintaining factor, and NO (81, 573, 631). Based on follicular dermis as early as 7 days PI (126). The lymphoid
infection of CEF with RB‐1B in the presence of IFN‐con- aggregates can develop into either necrotic areas consist-
taining supernatants, Levy et al. (340) suggested that IFN ing of FFE cells and degenerating lymphocytes or into
may block virus replication before translation of late cutaneous tumors. The former is associated with strong
genes. In one study IL‐10 expression leading to a Th2 expression of pp38, which is the first viral protein
response was the only cytokine upregulated in spleno- expressed in the FFE after reactivation followed by gB
cytes during latent infection (253), but this could not be and gD (398). In contrast, the cutaneous tumors have
confirmed in another study (422). only a few pp38+ cells. It is likely that virus is reactivated
Most latently infected cells are activated CD4+ T cells, from latency in the FFE, because mutant strains lacking
although CD8+ T cells and B cells can also be involved vIL‐8 (145) or pp38 (228) are able to produce virus in the
(105, 337, 521). Infection in genetically resistant birds FFE. Detailed dynamics of MDV interaction in the
often remains latent and can last for the lifetime of the feather and its importance in spread has recently been
bird aside from a persistent low‐grade productive infec- reviewed (249, 286).
tion in the FFE (90). Apoptosis of T cells during latent
infection has been described (378, 380), although it can-
Development of Lymphomas (Phase 4)
not be excluded that MDV was reactivated in these cells.
Susceptible birds or resistant birds infected with vv or Lymphoproliferative changes, constituting the ultimate
vv+ strains may develop a second wave of cytolytic infec- response in the disease, may progress to tumor develop-
tions after the second or third week, coincident with per- ment. Death from lymphomas may occur at any time
manent immunosuppression. from about three weeks onward. Regression of lesions
Chapter 15  Neoplastic Diseases 575

has been reported after infection with vMDV strains and vv+ strains may cause a prolonged and more severe cyto-
depends on the genetic resistance of the bird and the age lytic infection (642). Some of the new strains are capable
at infection (75, 323, 519). However, it is not clear how of infecting macrophages leading to increased death of
important tumor regression is in commercial poultry. macrophages (38). Murata et  al. (385) suggested that
The transformed T cells are mostly CD4+ cells expressing point mutations in meq resulting in amino acid substitu-
TCRαβ1 or TCRαβ2 and MHC‐II (495) with limited clonal- tions in the protein could change transactivation and
ity (386). Other subsets (e.g., CD8+CD4−, CD3−CD4−CD8− hence change the pathogenicity.
and CD3+CD4−CD8−) can be transformed under special The immune response itself may be responsible for
conditions (99, 402, 495). Burgess and Davison (77) further some lesions characteristic of MD. Nerve lesions have
characterized tumor cells using ex vivo lymphoma cells and some characteristics suggestive of an autoimmune dis-
tumor cell lines as MHC‐Ihi, MHC‐IIhi, CD4+, TCRαβ1+ or ease (449, 505), although the presence of MDV was not
TCRαβ2+, CD25+, CD28−, CD30hi. This profile is compati- positively shown. Additional evidence supporting an
ble with Treg cells (420, 511). The expression of CD30hi sug- autoimmune component for MD comes from studies
gests that MD could be a natural model for Hodgkin’s showing immune complexes in the kidneys of MDV‐
disease (78). Marek’s disease tumor cells may also express infected chickens and quail (306, 444).
poorly characterized MATSA (623) and fetal antigens (442). The primary cytolytic infection is not an absolute pre-
In addition to tumor cells CD8+ T cells are present and are requisite for tumor development. Schat et al. (491) found
consisting of oligoclonal expansions with public and private that MDV infection in EBx chickens resulted in the
CDR3 sequences (386). development of tumors in the absence of the primary
The infection in transformed cells is nonproductive in cytolytic infection, which also is the case in vaccinated
vivo and in vitro. CD4+ and CD30hi express Meq and chickens. However, stress and immunosuppressive infec-
SAR (see Viral Proteins), but are negative for pp38 and tions may induce secondary cytolytic infections, reduc-
gB (77, 477). Meq is the key protein involved in the trans- ing the benefits provided by vaccination.
formation of lymphocytes probably in conjunction with
viral telomerase (see Genes Unique for MDV) (388, 565). Pathogenesis of Non‐Tumor Diseases
The possibility that MD tumors are of clonal origin has Marek’s disease virus infection can cause several non‐
been proposed based on observations of random MDV neoplastic disease syndromes (Table 15.3B). The patho-
DNA integration into the genomes of lymphoma cells genesis of MDV‐induced atherosclerosis has not been
(162). Analyzing the TCRβ repertoire of tumors, Mwangi elucidated. Microscopic lesions consisting of fatty prolif-
et al. (386) concluded that most of the tumors are clonal erative lesions with alterations in lipid metabolism in
in origin, but that birds could have tumors originating arterial smooth muscle cells could be detected as early as
from different clones. In addition, PBL can be positive one month after infection (191, 237). In contrast to the
for these clonal tumor cells a few weeks before the birds original work by Fabricant et al. (192, 193), Njenga and
succomb. Earlier studies by Schat et al. (495) showed that Dangler (399) were unable to demonstrate arterial lipid
different lymphomas in the same bird could yield cell accumulation without cholesterol supplementation.
lines representing different T‐cell phenotypes. Cellular infiltrates were detected in the intima and serum
One of the major problems in understanding the cholesterol was increased significantly compared to non-
molecular basis for transformation has been the lack of a infected control chickens.
reliable method for in vitro transformation of T cells, The pathogenesis of the neurological lesion complex
however such a model has been published which will consisting of classical transient paralysis (TP), acute TP,
allow analysis of interaction between virus and target persistent neurological syndrome (PND), and late paral-
cells in an easily accessible system (503). ysis (LP) (231) is not fully understood. The difference
between classical and acute TP is somewhat arbitrary
Factors Influencing Pathogenesis (607) and the early pathogenesis is probably similar. The
The pathogenesis of infection with oncogenic MDV, development of both types of TP is influenced by the
which has become attenuated by passage in vitro, has MHC and the virulence of the MDV strain with the more
been studied by Bradley et al. (quoted in 427, 499) and virulent strains causing acute rather than classical TP
Schat et al. (494). Attenuated viruses failed to cause cyto- (504, 607). B cells are required for the induction of tran-
lytic infection and cell‐associated viremia levels were sient TP (418), probably because these cells are essential
low. Moreover, attenuated virus was not infectious for for the early cytolytic infection. The brain lesions start
lymphocytes in vitro, perhaps explaining the in vivo with vasculitis at 6–8 days followed by leakage of albu-
observations. min from blood vessels into vacuoles (550). This vaso-
Virus strains differ in oncogenicity, but the molecular genic edema is transient and correlates with classical TP
basis for differences in pathogenesis are not well defined. symptoms (548). Jarosinski et  al. (288) noted that the
All cause similar early cytolytic infections, although the development of neurological symptoms induced by
576 Section II  Viral Diseases

vv+RK‐1 correlated with increased levels of iNOS mRNA development. However, vaccination clearly reduces the
in the cerebellum and NO in blood serum (see Nitric early cytolytic infection (101, 252, 492), thus preventing
Oxide). Nitric oxide can cause vasodilation and could be extensive damage to the immune system and reducing
the cause of the edema. Chickens inoculated with the the number of latently infected T cells. Lesion regres-
vMDV JM‐16 strain did not show neurological signs or sion, however, has been described (75, 519) suggesting
iNOS mRNA in the cerebellum. that immune responses against tumor cells may occur.
The degree of virus replication in the brain may be
related to the severity of the disease. The absence or low Initiation of  Immune Responses.  Professional antigen‐
levels of virus replication correlated with the absence of processing cells (APC), such as dendritic cells, encountering
neurological symptoms (3, 229), while high levels of rep- pathogens are activated by interactions between the PAMP
lication in MHC resistant and susceptible chickens and the pattern recognition receptors (PRR), for example,
resulted in neural lesions (288). Attenuation of the TLR, on the APC. These interactions result in the activation
vv+648A strain resulted in reduced induction of TP of cytokines which direct both the innate and acquired
coordinately with a reduction in viral replication in lym- immune responses. There is currently no information on
phoid organs and FFE (229). PAMPs associated with any of the three MDV serotypes.
The clinical signs of PND are associated with a strong Although the distinction between innate and acquired
infiltration of lymphoblasts in the neuropil, many of immune responses has become less defined over the last
which express Meq protein. The later occurrence of PND few years, the innate and acquired responses will be dis-
after 3 weeks PI suggests that its pathogenesis may paral- cussed in separate sections.
lel that of lymphoma induction in other tissues.
Moreover, PND was shown to be closely related with the Innate Immune Responses.  Innate immune responses
onset of lymphoproliferative lesions in peripheral nerves include changes in cytokine expression, natural killer
and visceral organs (229). (NK) cells, and macrophages.

Cytokine Responses.  Infection with MDV results in the


Immunity
upregulation of a number of proinflammatory cytokines
Infection with pathogenic MDV or vaccine strains not (see Early Productive‐Restrictive Infection for details)
only results in the activation of innate, nonspecific and driving a TH1 immune response.
acquired, or specific immune responses but may also IFN‐γ is an important pleiotropic cytokine with many
cause immunosuppressive effects especially after infec- functions in antiviral immune responses, but few studies
tion with pathogenic serotype 1 strains. The importance have been performed on the roles of IFN‐γ in protective
of the interactions between immune responses and immunity to MD. In vitro studies indicate that IFN‐γ is
immunosuppression for the pathogenesis of MD cannot able to inhibit virus replication directly or indirectly
be overemphasized. A distortion in the balance toward through the induction of NO production and reactive
immunosuppression will lead to disease. Immune oxygen intermediates (166, 631).
responses and immunosuppressive features of MD have Detailed reviews on immunity to MD have been
been extensively reviewed (56, 157, 245, 497, 502). described elsewhere (56, 245, 246).

Immune Responses Nitric Oxide.  Nitric oxide is synthesized by three iso-


The immune responses developing during the early forms of NOS with iNOS (NOS II) being inducible in
cytolytic phase of infection are crucial for the outcome of macrophages, glial cells, astrocytes, and perhaps other
infection. Impairment of immune responses during this cells as well. The induction of iNOS occurs as part of the
phase delays establishment of latency resulting in pro- nonspecific inflammatory immune response to micro-
longing the lytic infection and the subsequent continued organisms. Nitric oxide and other reactive nitrogen spe-
destruction of immune cells by virus‐induced apoptosis. cies are very versatile molecules with many functions.
Impairments include infection at one day of age when Nitric oxide has been linked to beneficial affects by kill-
the immune responses are not yet fully developed or ing pathogens but also to neurodegenerative processes
treatment with cyclosporin or neonatal thymectomy in humans (65, 171).
combined with cyclophosphamide treatment (82). The Nitric oxide can inhibit MDV replication in vitro (166,
importance of immune responses during latency is rele- 631). Increased transcription of iNOS has been reported
vant for protection against the second cytolytic phase between 6–12 days PI with MDV (630) resulting in
and is dependent on CMI. It has often been suggested increased levels of NO in the plasma of genetically resist-
that vaccine‐induced immunity is an antitumor immune ant but not in genetically susceptible chickens (167, 290).
response because vaccination does not prevent superin- Nitric oxide may be beneficial, because it inhibited MDV
fection with wild‐type virus but does prevent tumor replication in vivo when genetically resistant chickens
Chapter 15  Neoplastic Diseases 577

were challenged with vMDV (631). However, pathology zation anti‐idiotype antibodies against MATSA (151).
may be associated with very high levels of NO produc- Similarly, antibodies against CD30 may provide protec-
tion especially in genetically resistant birds challenged tion against tumor cells (78).
with vv+MDV (288).
Cell‐Mediated Immunity.  Cytotoxic T lymphocytes (CTL)
NK Cells.  Natural killer cells are the first line of defense recognize small peptide fragments of 8–12 amino acids
because these cells can lyse virus‐infected and tumor presented in the context of MHC‐I antigens. These
cells without prior exposure to the pathogen. Natural peptides are generated from de novo synthesized proteins
killer cells are also potent inducers of IFN‐γ. In order to through a complex process involving the proteasome
lyse target cells, NK cells must recognize the target cells and transporters associated with antigen‐processing
as foreign (e.g., the MHC‐I has been altered or down- (TAP) 1 and 2. In vitro demonstration of antigen‐specific
regulated). Thus the downregulation of MHC‐I during CTL requires effector and target cells expressing the
the lytic infection (268) supports a potential role for NK same MHC‐I antigens (515).
cells. Thus far functional NK cell assays have used total Recently, Mwangi et al. (386) found a limited number of
spleen cell populations and LSCC‐RP9 as target cells in CD8+ clonal cell populations, which were generated
chromium release assays (CRA). The recent develop- shortly after infection suggesting that MDV infection
ment of NK cell‐specific mAb (284) will facilitate further results in a limited number of antigen‐specific clones.
examination of the importance of NK cells in MD Earlier, Pratt et al. (448) stably transfected and expressed
immunity. MDV genes in REV‐transformed cell lines with known
MHC antigens. These cell lines were used to show that
Macrophages. Activated macrophages can restrict CTL from infected or vaccinated chickens recognize pep-
MDV replication and perhaps as a consequence reduce tides derived from pp38, Meq, ICP4, ICP27, gB, gC, gH,
tumor incidence. These effects are probably the result of gI, and gE (361, 404, 502). The effector cells developed
NO production (497, 502) or by reactive oxygen inter- around 7 days PI and were characterized as typical CTL
mediates (166). Increased numbers of macrophages have expressing CD3, CD8, and TCRαβ1 but not CD4 (405).
been noted shortly after infection in the lung in conjunc- Important differences were noted in the recognition of
tion with increased transcription of iNOS. Macrophages proteins by CTL from resistant and susceptible chicken
harvested shortly after MDV infection can inhibit prolif- lines. Cytotoxic T lymphocytes from resistant N2a (MHC:
eration of MD cell lines in vitro, which was considered to B21B21) but not from susceptible P2a (MHC: B19B19)
be a transient immunosuppressive effect (334, 513). This chickens recognized ICP4 (404). Effective killing of
inhibition may actually be a protective response, because infected cells as soon as ICP4 is expressed, for example,
it may limit the number of activated T cells during the when latently infected cells are reactivated, and before
critical switch of MDV from B to T cells (497, 502). virus replication is completed could be one of the con-
tributing factors to MHC‐based genetic resistance.
Acquired Immunity Cytotoxic T lymphocyte‐specific responses to gB, gI and
Humoral Immunity.  Chickens infected with MDV pp38 also appear important to MD protection. (328, 394).
develop precipitating and VN antibodies within 1–2 Cytotoxic T lymphocytes and NK cells can lyse target
weeks; a transient IgM response is replaced by IgY (256). cells through the perforin/granzyme pathway. Increased
Due to the cell‐associated nature of MDV, antibodies are transcription rates of Granzyme A occur between 4–15
of limited importance in MD immunity. Virus‐ days PI (251, 480, 481).
neutralizing antibodies are important only when cell‐
free virus infects chickens or when MDV proteins are Vaccinal Immunity.  Herpesvirus of turkey, attenuated
expressed on the surface of cells. In the latter case, MDV, and serotype 2 MDV protect against early
antibodies plus complement or antibody‐dependent, replication of virulent viruses in the lymphoid organs of
cell‐mediated cytotoxicity (ADCC) can lyse infected challenged birds, reduce the level of latent infection, but
cells (317, 472). However, the target antigens and the do not prevent infection (reviewed in 211, 497, 600).
effector cells involved in ADCC were not identified. In Based on current knowledge, the following sequence of
vivo VN has indeed been demonstrated using cell‐free events is proposed to explain vaccine‐induced immunity
and cell‐associated virus (79). Maternal antibodies with challenge occurring within three days after hatch as
reduce the cytolytic infection (86) and can reduce the is typical in the field (Figure 15.14).
efficacy of cell‐associated vaccines with low titers or if Vaccination with CVI988 results in upregulation of
cell‐free HVT is used (111, 315). IFN‐γ, IL‐8, IL‐18, and iNOS in lung and spleen as early
The possibility that surface antigens found on MDV‐ as 3 days PI (213), which is probably also the case after
transformed cells could be involved in immunity was vaccination with HVT and SB‐1. IFN‐γ can reduce virus
suggested by protection against challenge after immuni- replication and stimulate macrophages to initiate the
578 Section II  Viral Diseases

transcription of iNOS, producing NO between 3 and 7 concurrently and, in such cases, immunosuppression
days post vaccination, thus limiting the replication of may serve to augment oncogenic potential.
challenge virus. Shortly afterwards, NK cells are acti- Humoral and CMI can be suppressed by MDV infec-
vated (254, 514), probably producing more IFN‐γ and tion leading to reduced antibody responses to a variety of
killing virus‐infected B cells. antigens and alterations in T cell functions, such as skin
Feather pulp can be used to examine temporal changes graft rejection, mitogen stimulation of lymphocytes,
in immune responses in individual, vaccinated chickens. delayed hypersensitivity, reduced NK cell activity, pri-
This will facilitate monitoring cytokine responses mary and secondary infections with coccidia, and
induced by different vaccines (5, 244). Vaccination with impaired Rous sarcoma regression (citations in 427, 499).
CVI988 showed a down regulation of RB‐1B in the
feather pulp (244). Challenge did not increase replication
of CVI988 (34, 244) in contrast to the observation that Diagnosis
HVT and SB‐1 shedding increased after challenge with
MDV (278). Techniques for diagnosis of infection with MDV are dif-
Immunoevasion, caused by MDV, can interfere with ferent from those needed for differential diagnosis of the
vaccine‐induced immunity. MHC‐I downregulation is disease. The infection is ubiquitous, but the disease is
one strategy to reduce the host defenses, as reviewed not. The principal methods to identify the presence of
elsewhere (245). The MDV gene MDV012 was recently infection are isolation of the virus, demonstration of viral
demonstrated as capable of reducing surface expression DNA or antigens in tissues, and detection of antibody.
of MHC‐I on chicken cells (248). The host response can The applications of different diagnostic procedures have
also be altered by differences between vaccines from dif- been recently reviewed (223, 401, 606).
ferent manufacturers, which probably relates to passage
levels (331). Concurrent infections with immunosup-
Virus Isolation
pressive viruses, for example, CIAV (362) or stress may
interfere with vaccine‐induced CMI responses. Deletion Virus isolation is performed to confirm its presence for
of humoral immunity by bursectomy and X‐irradiation diagnostic purposes and to secure the infectious virus
does not seem to have a major effect on protection con- for further study. Techniques for isolation of all sero-
ferred by attenuated MDV (183), although similar treat- types have been reviewed (223, 487).
ment partially impairs HVT vaccine immunity (467).
Source of Virus
Immunosuppression Marek’s disease virus can be isolated as early as 1 or 2 days
Suppression of the immune response by MDV infection PI or 5 days after contact exposure and throughout the life
is a critical feature of the disease, contributing to the of the chicken (reviewed in 499). Intact viable cells are the
virulence of MDV isolates and altering susceptibility of preferred inoculum because, in most cases, infectivity is
the host to other pathogens (reviewed in 486, 500). Initial avidly cell‐associated, although cell‐free preparations from
impairment of the immune response is the result of the skin, dander, or feather tips of infected chickens may con-
lytic infection of lymphocytes during the first cytolytic tain the virus (92). Inocula may consist of blood lympho-
infection (see Pathogenesis) (citations in 486, 500). The cytes, heparinized whole blood, splenocytes, or tumor cells.
onset of immunosuppression later in life appears to be a Marek’s disease virus can often be recovered from infected
unique feature of vv+MDV strains and is not eliminated cell suspensions following s­ torage for 24 hours at 4°C, thus
by vaccination (196, 197). It is difficult to distinguish facilitating transport of samples (reviewed in 499).
between cause and effect because tumor cells might have
suppressor activity (69, 456). Because immunocompe- Cell Culture Techniques
tence is required for the maintenance of latency (82) it Probably, the most widely used method for primary iso-
might be that immunosuppression associated with the lation of MDV is inoculation of susceptible cell cultures
appearance of transformed lymphoblasts results in addi- with blood lymphocytes or single‐cell suspensions from
tional reactivation of the lytic infection. This, in turn, lymphoid tissues of infected chickens. Chicken kidney
will cause the loss of additional B and T cells, thus com- cell and DEF cultures are preferred substrates for ­primary
pounding the situation and resulting in the bursal and isolation of serotype 1 MDV; whereas CEF normally are
thymic atrophy seen in birds destined to succumb to used for isolation of viruses of serotypes 2 and 3 as well
MD. However, immunosuppression may not be a prereq- as for attenuated serotype 1 vaccine strains. Although
uisite for the development of tumors, as observed with CEF are less permissive for growth of low passage
several experimental vaccine candidates (175, 611). ­serotype 1 virus (reviewed in 487), some contemporary
Although virus‐induced immunosuppression and onco- isolates may grow well in CEF, even on primary isolation.
genicity are not invariably linked, they are often expressed Cultures are inoculated with 1–2 × 106 cells, although
Chapter 15  Neoplastic Diseases 579

some inhibition of viral plaque formation may be Such infection markers also have value for the identifica-
encountered with doses greater than 8 × 106 cells for tion of putative MDV isolates in cell cultures. However,
some viruses (109). only detection of oncogene meq and high load of MDV
Development of typical plaques (Figure 15.3) in inocu- DNA in tumors can be used as diagnostic criteria for the
lated cultures within 3–12 days and the absence of such disease (226).
changes in comparable uninoculated (or sham inoculated)
control cultures are evidence for isolation of MDV. The Viral Antigen Detection
plaques induced by serotype 1, 2, and 3 viruses can be dis- Monoclonal antibodies prepared against type‐common
tinguished, with practice, by morphologic criteria (484, and type‐specific epitopes of all three MDV serotypes
590), but IF staining with serotype‐specific mAbs provides (332) are now used in preference to polyclonal antibodies
a more accurate differentiation. Optimal time for observa- for the detection of antigens in tissues. Monoclonal anti-
tion of plaques varies with the cell substrate and serotype bodies H19 and T65 can be used to differentiate CVI988
of the virus. Marek’s disease virus also has been isolated by from other serotype 1 MDVs strains based on differences
direct culture of kidney cells from infected chickens or by in the amino acid sequence of protein pp38 (147, 220).
inoculation of normal kidney cultures with trypsinized Viral antigens can be detected in feather tips and FFE,
kidney cells from infected chickens (621). cytolytically infected lymphoid tissues, brain, or infected
cell cultures with appropriate antibodies by fluorescent
Isolate Identification antibody tests, immunohistochemistry, dot‐ELISA (322),
Marek’s disease virus serotype 1 isolates should be free of agar gel precipitin (AGP) tests, and immunoassay
contaminating MD vaccine strains. It normally is useful (reviewed in 223, 499). In MD lymphomas, oncogene meq
for the isolate to be plaque purified or cloned at the earli- is the only antigen that is consistently expressed (226, 477)
est possible passage. Serotype identity and purity can be but pp38‐positive cells occasionally are observed (226).
confirmed using staining techniques with serotype‐spe-
cific mAbs (332). Freedom from extraneous viruses is Viral Nucleic Acid Detection
critical, because contamination with passenger viruses Polymerase Chain Reaction (PCR) Assay.  The availability of
may alter the apparent pathogenicity of the isolate (291, the nucleotide sequences of different genes from a large
409). Propagation of MDV isolates for up to six passages number of viruses including the complete genome
in CEFs or CKC cultures appears to exclude contaminants sequence of the three serotypes of MDV allows the use of
such as CIAV (641) and permits preparation of seed and PCR‐based methods of specific detection of MDV.
working stocks, which can be more easily standardized Primers designed to amplify sequences specific for each
and titrated. To preserve virulence, some workers have serotype (277, 464) as well as to differentiate between
preferred to propagate serotype 1 viruses in vivo, prepar- serotype 1 oncogenic and attenuated strains based on the
ing stocks of cryopreserved spleen or buffy coat cells from 132 bp (42, 525, 655), and between CVI988 and other
infected chickens. Pathotyping of serotype 1 MDVs, serotype 1 MDV strains (30, 220, 468) have been
although not routine, may be accomplished by compari- described. However, PCR may not always be sensitive
son of pathogenicity with that of prototype strains by enough to detect latent infection due to the lower
inoculation of non‐vaccinated chickens as well as chickens frequency of positive cells and the lower number of viral
vaccinated with HVT or bivalent vaccines (604). genomes per cell. Furthermore, detection of MDV DNA,
even from an oncogenic MDV, does not have any
Virus Assay and Titration diagnostic value as chickens get exposed to oncogenic
Viruses of serotypes 1, 2, and 3 can be assayed by in vitro MDVs, if properly immunized, they never develop MD.
techniques similar to those described for virus isolation. Quantitative PCR (qPCR) assays using various primer
Methods differ for different serotypes, but most rely on sequences have been used to assay viral load in tissues
plaque induction in susceptible cell cultures. Enumeration from infected chickens (30, 31, 72, 76, 220, 277, 464) and
should be done as soon as plaques become mature (time are essential tools for diagnosis and epidemiological
varies with isolate), because secondary plaques may studies of MD. Unlike conventional PCRs, qPCR has
occur when cultures are maintained with liquid medium. been shown to be very valuable in the diagnosis of MD
Procedures for titration of vaccine viruses have been (218, 220, 226, 368, 582), for monitoring MD vaccination
reviewed (559) and are not fundamentally different from (136, 214, 216, 217, 220, 241, 276, 279, 469), and for stud-
those for pathogenic isolates. ying different aspects of MD biology including replica-
tion kinetics of MDV in blood, feathers, lymphoid
tissues, and dust (34, 136, 244, 276, 279, 280, 457–459).
Viral Markers in Tissues
It is often desirable to detect the presence of viral infec- DNA Probes.  Methods using DNA–DNA dot‐blot
tion in chickens without isolating the virus in culture. hybridization with DNA probes for the detection of
580 Section II  Viral Diseases

MDV DNA in feather tip extracts (154) have been used ­ rovisionally as MD if at least one of the following con-
p
for detection of MDV (382–384). Furthermore, ditions is met: (1) leukotic enlargement of peripheral
localization of virus‐infected cells has been accomplished nerves; (2) lymphoid tumors in various tissues (liver,
by in situ hybridization for both MDV (185, 434, 475) heart, gonad, skin, muscle, and proventriculus) in birds
and HVT (265) and by fluorescence in situ hybridization under 16 weeks of age; (3) visceral lymphoid tumors in
(FISH) for MDV (302). birds 16 weeks or older that lack neoplastic involvement
of the bursa of Fabricius; or (4) iris discoloration and
Loop‐Mediated Isothermal Amplification.  Loop‐mediated pupil irregularity, as in Figure  15.8C. Proper examina-
isothermal amplification (LAMP) methods for the rapid tion of the bursa is particularly important and requires
detection of MDV‐1 genome at high sensitivity were incision of the organ with close inspection of the epithe-
developed for detection of MDV in less‐equipped lial surface. However, diagnoses based only on gross
laboratories as well as under field conditions (17, 175, pathologic criteria are not definitive and additional
625, 627, 628). steps are required.

Step 2—Histology, Cytology, and Histochemistry


Antibody Detection
of Tumor Cells
Tests for identifying the presence of specific antibodies Affected tissues, fixed in formalin or fresh‐frozen, are
in chicken sera are useful in studies of viral pathogenesis used to prepare paraffin and cryostat sections, respec-
and for monitoring SPF flocks. However, it does not have tively. Impression smears of tumors may also be used
any value in the diagnosis of MD in commercial flocks. A (524). Histopathology can be very useful to confirm the
number of procedures including AGP, fluorescent anti- diagnosis of lymphoma and to evaluate the morphology
body ELISAs (373, 646), and VN tests are in common use and distribution of tumor cells. Marek’s disease tumors
(review in 223). consist of a mixed population of small to large lympho-
cytes, lymphoblasts, plasma cells, and macrophages
(222, 426). The presence of such infiltrates in the nerves
Diagnosis of the Disease
(lesion type A) is the only pathognomonic lesion of MD
Despite long‐established guidelines for the pathologic (326, 426). However, MDV is also able to induce inflam-
diagnosis of MD (524), diagnosis of the clinical disease matory lesions characterized by edema, demyelination,
remains difficult in practice because of the absence of and plasma cell infiltration (lesion type B) that are not
truly pathognomonic gross lesions and the widespread necessarily related with MD lymphoma and can be con-
nature, often with coinfection, of other pathogens such fused with peripheral neuropathy (25, 228, 326, 605).
as ALV and REV (149, 153), complicating diagnostic Likewise, minor infiltration of lymphocytes in peripheral
efforts that depend on virological methods. nerves might be indicative of infection with MDV but
Diagnosis of MD must primarily be addressed by con- not of MD lymphoma (review in 606).
sideration of the characteristics of the proliferating cell Characterization of the cell phenotype by immunohis-
populations that constitute the disease. Other disease‐ tochemistry can aid in the differential diagnosis of MD
specific criteria, such as epidemiological factors, are also lymphomas. Marek’s disease tumor cells express MHC
valuable. Detection of virus, viral DNA, or viral antigens class II antigen and T‐cell markers, especially CD4 (495).
(with the exception of oncogene meq) does not have any CD30 (477) and MATSA (623) are commonly found in
diagnostic value since infection is ubiquitous and vacci- MD tumor cells although they are also present on retro-
nation does not prevent superinfection (226). However, virus‐induced B cell lymphoma (226) and activated T
quantification of MDV DNA load in tumors is a diagnos- cells (226, 365).
tic criterion that can confirm the diagnosis of MD (218,
220, 226). The process commences with the acquisition of Step 3—Virologic Criteria
a flock history and a sufficient number (5 to 10) of repre- For tumors that satisfy MD criteria listed in steps 1 and 2
sentative sick and dead chickens showing the lesions of or for atypical tumors, the association of MDV with the
the disease and proceeds as a series of steps (606). tumor cell is a useful confirmation. Viral antigen Meq
can be consistently detected in tumor cells by in situ
Step 1—Clinical Data and Gross Pathology hybridization, immunohistochemistry, or fluorescent
Although enlarged peripheral nerves and visceral lym- antibody tests and can be used as diagnosis criterion to
phomas are common in MD and one or both are invari- confirm MD tumors (226, 477). Detection of other viral
ably present, neither lesion occurs consistently nor is antigens such as pp38 has been described in tumors
pathognomonic. Thus, other criteria, such as age and (390) but expression is sporadic, it seems to be related to
lesion distribution, must be considered in the postmor- reactivation of virus in neoplastic cell, and it is not an
tem diagnosis of MD. Chickens may be diagnosed adequate diagnostic criterion for MD tumors (226).
Chapter 15  Neoplastic Diseases 581

Polymerase chain reaction assays and virus isolation REV in chickens older than 16 weeks of age. Chickens
from buffy coat, spleen cells, or feather pulp samples as usually have gross tumors in the bursa of Fabricius, and
well as detection of viral proteins demonstrate the virus tumor cells are uniform, blast‐like, pyroninophilic, and
in the bird but do little to associate the virus with the express B‐cell markers and IgM. Also, the tumor cells
tumor cells. However, there is a quantitative association have clonal insertions of proviral DNA near the c‐myc
between virus load and MD tumors. Low levels of virus gene (see Leukosis/Sarcoma Group). Nerve enlarge-
or viral DNA may be detected in lymphocytes from ment, runting, and nonbursal T‐cell lymphomas can be
nontumor‐bearing chickens, but most tumor‐bearing
­ induced by REV but, thus far, have only been observed
chickens have high viremia titers (622) and high load of under experimental conditions or where chickens
MDV DNA measured by qPCR (33, 218, 220, 226, 459). have  been inoculated with contaminated vaccines.
Thus, the demonstration of high load of MDV DNA in Lymphocytes obtained from REV‐induced nerve lesions
tumor cells and the absence of other tumor viruses, along or tumors do not express Meq and have low MDV DNA
with the criteria in steps 1 and 2, is sufficient to establish load, if any (226). Cells from nonbursal RE lymphomas
MD diagnosis. Evaluation of MDV DNA load in blood of are negative for MHC class II and predominantly stain
feather pulp of chickens as early as 21 days of age can also for CD8 antigen (606) (see Reticuloendotheliosis).
be used to predict the MD outcome on those flocks (218, Exclusion of ALVs or REVs, where possible, through neg-
220, 459). Proper collection of samples for MD diagnosis ative PCR, histochemical assays on tumors, or antibody
using qPCR has been recently reviewed (223). tests may provide strong support for a diagnosis of MD
when other MD‐related criteria are positive. However, it
is possible to have mixed infections and in those cases
Pathotyping of MDV Strains
further diagnostic assays (qPCR for MD and southern
The concept of MDV pathotypes has arisen from the blot for retroviruses) are needed (190, 226).
recognition of the existence of strains that are associated Peripheral neuropathy is a neurological disease of
with increased virulence that show correlation with uncertain etiology that causes paralysis and nerve
breaking of vaccinal immunity in the field (604). The enlargement in a low proportion of commercial chickens
ADOL (Avian Disease & Oncology Laboratory) method 6–12 weeks of age (25, 53) and is characterized by a Th1‐
of pathotyping, based on induction of lymphoprolifera- to‐Th2 shift (26). Affected chickens lack visceral lym-
tive lesions in chickens vaccinated with different vacci- phomas; the nerve lesions are uniformly B‐type; and
nation regimes is the most widely used. This method was MDV is rarely, if ever, demonstrated. If MDV is present
used to characterize more than 45 isolates into distinct on those chickens, MDV DNA load is low (606). Other
vMDV, vvMDV, or vv+MDV pathotype groups (597). diseases that may present confusing gross lesions or par-
Even though the ADOL method stipulates the use of line alytic signs are myelocytomatosis (myeloid leukosis),
15x7 chickens for pathotyping, experiments with other myeloblastosis, erythroblastosis, histiocytic sarcomas,
lines of birds have given similar results (83, 170, 604). carcinoma of the ovary, various other nonviral neo-
The ADOL pathotyping assays are cumbersome and plasms, riboflavin deficiency, tuberculosis, histomonia-
require infrastructure that is lacking in most laborato- sis, genetic gray eye, Newcastle disease, hepatitis E, and
ries. Therefore, different methods to differentiate joint infections or injuries. Myeloid leukosis is a com-
between classical strains of MDV and the more virulent mon tumor in broiler breeder flocks that superficially
pathotypes have been examined including neuropatho- resembles MD but can be differentiated histologically.
typing (230), virus replication (172), lymphoid organ The tumor cells are myeloid in nature and lack T cell and
atrophy and immunosuppression (95, 196), and sequence MD viral markers.
of various genes (512, 555). However, classification
obtained by the alternative methods do not always ren-
Diagnosis of Other MD Syndromes
der the same results as ADOL pathotyping and most of
these techniques are considered an adjunct to pathotyp- Transient paralysis occasionally is observed in the field,
ing more than substitutes of the “gold standard” ADOL especially in chickens not vaccinated against MD once
pathotyping assay. maternal antibodies have waned between 30–40 days of
age (reviewed in 222). Development of TP in vaccinated
commercial flocks would indicate failures on the vacci-
Differential Diagnosis
nation process as proper vaccination against MD pro-
Details on how to do differential diagnosis of MD with tects against the development of TP (reviewed in 222).
other poultry tumor diseases has been reviewed (606). Diagnosis of TP is done based on the history (sudden
The major differential diagnosis for MD is lymphoid leu- onset of flaccid paralysis that last 24–48 hours and can
kosis (LL). Lymphoid leukosis is a clonal, bursal lym- lead to death or total recovery), lack of gross lesions in
phoma induced by ALV and, under some conditions, by nerves or viscera, and histopathology (brain edema and
582 Section II  Viral Diseases

vasculitis). There are three diseases that need to be of MD. Genetic resistance and biosecurity, however, are
included in the differential diagnosis with TP: botulism, critical adjuncts to properly executed vaccination proce-
neurological form of MD (fowl paralysis), and peripheral dures. No effective practical treatment exists for the
neuropathy. Both TP and botulism cause flaccid paraly- ­disease in individual chickens or infected flocks. An inte-
sis of the neck progressing to the limbs and are not asso- grated strategy to prevent early infection, to slow the
ciated with gross lesions in the nerves or viscera. acquisition of virulence of field strains, and to provide
Confirmation of TP can be done by histopathological superior immune responses seems most likely to suc-
examination of the brain as botulism does not produce ceed. Various reviews on MD vaccines and control
brain edema and vasculitis characteristic of TP. Unlike ­procedures are available (67, 211, 462, 489).
TP, the neurological form of MD appears as permanent
spastic paralysis with enlargement of peripheral nerves
Vaccination
and pathognomonic type A lesions in the nerves.
Peripheral neuropathy tend to last longer than TP and is Types of Vaccines
also characterized by peripheral nerve enlargement with Several different types of MD vaccines are in common
lesions that resemble MD B‐type lesions. Davidson et al. use, both individually and in various combinations. The
(155) differentiated transient paralysis from peripheral most widely‐used products are low pathogenic serotype
neuropathy on the basis of PCR tests for MDV on brain 1 MDV attenuated in cell culture (470) and naturally
tissue. However, brains from MDV‐infected chickens nononcogenic serotype 3 (HVT) (403), and serotype 2
without transient paralysis may also be detected as posi- viruses (490, 618). The latter usually are combined with
tive by PCR assays (607). In contrast, detection of viral HVT to take advantage of the synergistic activity
antigens in the brain appeared to correlate with the onset ­documented between serotypes 2 and 3 (492, 589). All
of paralytic signs (227). vaccine types are protective but to varying degrees.
Skin leukosis (the skin form of MD) can be differenti- Herpesvirus of turkey virus, mainly strain FC126 (613)
ated from dermal squamous cell carcinoma, which is is extensively used because it is effective and economical
commonly observed in defeathered broiler chickens at to produce and combines well with other products.
processing (236, 391). Marek’s disease lesions are nodu- Although both cell‐free and cell‐associated forms of
lar and contain lymphoid cells, whereas squamous cell HVT are available, the latter has been most widely used
carcinomas have a craterlike gross appearance and are because it is more effective than cell‐free virus in the
composed of squamous epithelial cells. presence of maternal antibodies (603). Bivalent vaccines
Due to its complexity, the most difficult syndrome to consisting of HVT and SB‐1 (490) or 301B/1 (618)
diagnose is MDV‐induced immunosuppression (MDV‐IS). strains of serotype 2 MDV were introduced in the mid‐
Early MDV‐IS is associated with replication of MDV in the 1980s. The CVI988 strain (470) used in the Netherlands
lymphoid organs of unvaccinated chickens lacking mater- (356) and other countries since the early 1970s, was
nal antibodies and it is characterized by atrophy of the bursa introduced to the United States in the early 1990s.
and thymus. However, under field conditions lymphoid Serotype 1 and 2 vaccines are available only as cell‐­
organ atrophy could be due to several infectious and nonin- associated products.
fectious diseases. Experimentally it is possible to detect Besides the traditional vaccines, in the recent years a
MDV antigens in the lymphoid organs before the atrophy number of recombinant vaccines have been developed,
occurs, however such expression occur for a short period licensed, and are currently commercialized (reviewed in
(1–2) days and is not associated with clinical signs or gross 462). Efforts to develop recombinant MD vaccines started
lesions. Late MDV‐IS is even more complicated to diagnose in the early 1990s using either fowlpox virus (394) or
than early MDV‐IS as it is not associated with lymphoid HVT (374, 474) as vectors. However, it has been in the
organ atrophy and/or tumors (195–197). Because late‐ last few years that several products using HVT as a vector
MDV‐IS cannot be p ­ rotected by currently vaccination pro- for various diseases (infectious laryngotracheitis, infec-
tocols (197), it is very likely causing problems in commercial tious bursal disease, Newcastle disease, and avian influ-
flocks and should be considered in the differential diagnosis enza) have been licensed and widespread used; either
when immunosuppression is suspected. alone or in combination with MD vaccines of other sero-
types (188, 189, 205, 208, 232, 292, 339, 411, 435, 568). In
addition to HVT vector vaccines, recombinant vaccines
Intervention Strategies based on serotype 1 MDV strains have been developed
either by using attenuated serotype 1 MDV strains as vec-
The development of successful vaccines for control of tor (275, 349, 566, 647), by modifying or deleting genes
MD (132, 403, 470, 488) was a significant achievement. (62, 144, 333), by insertional mutation of the long termi-
Vaccination represents, for now and the foreseeable nal repeat (LTR) region of the REV in various MDV sero-
future, the central strategy for the prevention and ­control type 1 strains (149, 313, 353, 364, 611), or by combining
Chapter 15  Neoplastic Diseases 583

various techniques (i.e., REV LTR insertion and deletion significantly reduced in broilers. The older literature
of meq, meq‐deleted vaccines as vectors) (215, 545). To suggests that increased doses of HVT above a threshold
date, the only recombinant vaccine based on serotype 1 level of around 400 PFU did offer little improvement
MDV strains that is licensed and commercialized in some (178). However, recent studies indicate that a substan-
countries is a recombinant CVI988 vaccine with an inser- tially higher threshold is needed to protect against chal-
tion of the REV LTR (66, 353). This vaccine has been lenge with vv and vv+ strains (214, 216, 217).
shown to replicate and protect better than the parental Revaccination has been recently shown to increase
CVI988 and does not induce lymphoid organ atrophy in protection against early challenge with MDV (219, 224,
susceptible chickens lacking maternal antibodies (66). 629). The benefits of double vaccination has been repro-
However, experimental vaccine based on vvMDV strain duced under laboratory conditions (224) when the sec-
Md5 with deletion of both copies of the oncogene meq ond vaccine is more protective than the first vaccine
(352, 526) has been shown to be the most protective vac- administered and both vaccinations occur before chal-
cine against early challenge with vv+MDV (115, 331). lenge with oncogenic MDV (224). It has been suggested
This vaccine protects not only against the development of that the best revaccination program will include a low
tumors but also against the development of late MDV‐IS protective vaccine in ovo (i.e., HVT) followed by a high
in commercial meat type chickens (197). Furthermore, it protective vaccine at day of age (i.e., HVT+SB‐1 or/and
can be successfully used as a vector vaccine for other CVI988) (219).
poultry diseases (215, 651). Unfortunately, the deletion Several other factors may influence vaccine efficacy
mutant is causing severe thymus and bursa atrophy in such as the passage level of the vaccine viruses. Increased
maternal antibody‐negative, one‐day‐old chicks (175) passage level may cause a decrease in vaccine replication
preventing licensing in the United States under current resulting in decreased immunity (330, 614). Unfortunately,
regulations. The negative effect on the lymphoid organ in passage levels of commercial vaccines are in general not
chickens lacking maternal antibodies can be reversed by available. Maternal antibodies against the three serotypes
attenuation in cell culture or by adding a UL5 helicase‐ are typically present in commercial one‐day‐old chicks.
primase subunit point mutation; albeit at a cost of reduc- These antibodies may reduce the effectiveness of cell‐
ing protection (257, 330). associated vaccines but do not abrogate the protective
Other strategies to improve vaccines through recom- effect (315), if sufficient PFU are administered.
binant DNA approaches that might have potential use in In addition to improper vaccination techniques (381),
the future includes DNA vaccines (563), incorporation of early exposure is undoubtedly one of the most important
cytokines (168, 243, 554), and Toll‐like receptor‐based causes of excessive MD in vaccinated flocks because field
adjuvants (419, 421). exposure usually occurs very soon after placement of
chickens (612) and because at least seven days is required
Vaccine Administration to establish solid immunity after vaccination (281)
Marek’s disease vaccines are administered to chicks at (reviewed in 502). The vaccine strain of virus also has a
hatch by subcutaneous or intramuscular inoculation or major influence on vaccine efficacy. Immunity induced
in ovo at ED18 (67, 516) In ovo vaccination is now per- by weaker vaccines such as HVT may be excellent against
formed by automated technology and is used in more low virulence challenge but can be completely over-
than 90% of commercial broiler chickens in the United whelmed by early challenge with highly virulent strains
States (586). In ovo vaccination not only reduces labor (597). Protection conferred by CVI988, however, does
costs and has greater precision of vaccine administration not seem to be as affected by the pathotype of the
but also confers better protection against early challenge ­challenge strain (459) as vaccines of other serotypes.
with MDV than one‐day‐old vaccination (649). Although high virulence strains commonly are invoked
Furthermore, it has been recently shown that in ovo vac- to explain field outbreaks of disease, many alternate
cination with HVT hastens the maturation of the causes should be considered.
immune system of the chicken embryo rendering chicks Stress appears to interfere with the maintenance of
more immunocompetent at hatch (221). Deposition of vaccinal immunity. In chickens properly vaccinated at
the vaccine by the amniotic or intraembryonic route is hatch and well‐protected following challenge, Powell
essential for optimal protection (580, 585). Proper han- and Davison (441) induced MD lesions and mortality
dling of vaccine during thawing and reconstitution is by immunosuppressive treatment at 10 weeks of age. The
crucial to ensure that adequate doses are administered onset of egg production deserves consideration as a stress
(207, 239). factor precipitating vaccine breaks (600). Infection with
other pathogens such as IBDV, REV, CIAV, and r­ eoviruses
Factors Affecting Efficacy have been reported to interfere with the induction of vac-
Vaccines typically are given at doses of 2,000–6,000 cinal immunity (reviewed in 500), although very specific
plaque forming units (PFU) per chick, but these are often conditions are sometimes required. T2‐toxin has been
584 Section II  Viral Diseases

recently shown to have detrimental effect on the MD current MDV isolates are still vv+MDV strains suggest-
outcome of nonvaccinated chickens but it did not affect ing that once vv+MDV infection gets established in a
protection conferred by HVT (321). farm it will stay (173).
The strain of chicken is also an important determinant The emergence of increasingly virulent viral strains,
of vaccine efficacy. Schat et  al. (492) found that HVT coupled with an apparent reduction of vaccine efficacy
vaccine in genetically resistant chickens resulted in a during the past 20 years, has prompted justifiable con-
stronger immunity than did the bivalent (HVT+SB‐1) cern. This suggests that vaccination by itself does not
vaccine in susceptible chickens. Chang et  al. recently provide a complete control program and is not the ulti-
demonstrated that in genetically resistant lines of chick- mate solution for MD. In fact, recent evidence also points
ens, HVT can induce as much protection as some of the towards the potential contribution of the early genera-
commercial CVI988 strains (117). In commercial chick- tions of vaccines themselves driving virulence and selec-
ens, protective immunity conferred by vaccines is influ- tion of more virulent MDV pathotypes (461). Strict
enced by the B‐haplotype (22) as well as by non‐MHC biosecurity procedures to reduce early exposure and the
genetic variation (116). presence of genetic resistance are essential adjuncts to a
successful vaccination program. Furthermore other than
Vaccination Strategies increased protection against tumors vaccine research
Marek’s disease vaccines as a class are unusually effec- should focus on protecting against other aspects of MDV
tive, often achieving greater than 90% protection under infection such as infection, transmission, and MDV‐IS.
commercial conditions (600). However, attention is often
focused on flocks in which MD losses are perceived to be Genetic Resistance
excessive (381). Causes for such vaccine failures are dif- The well‐known variation in susceptibility of different
ficult to ascertain by retrospective analysis (381, 600). lines of chickens to MD challenge is determined by
However, there are several checkpoints that could be genetic factors (20, 71, 120) and provides a unique
evaluated in the event of an immunization failure (212). opportunity to include genetic approaches to control
Auditing at the hatchery to evaluate improper vaccina- MD. Indeed, poultry breeders have included resistance
tion techniques, titration of the batch of vaccines used, to MD in selection programs for many years (366).
replication of the vaccine in the chickens, early exposure However, genetic resistance can be overcome by chal-
to oncogenic MDV, coinfection with other immunosup- lenge with highly virulent MDVs and is best applied in
pression agents (especially with CIAV infection), and the concert with vaccination and biosecurity to achieve opti-
emergence of new MDV strains with increased virulence mal control. Genetics influences virtually every aspect of
need to be considered (212). host response to MD. However, only those issues ger-
Efficacy data comparing certain groups of vaccines are mane to disease control programs are considered here.
available (citations in 485, 600). Bivalent serotype 2+3 For successful incorporation of selection for resistance
vaccines are clearly more effective than HVT, but the in breeding programs several conditions need to be met
original CVI988 vaccine seems to be the most effective (reviewed in 20). The heritability of resistance is rela-
(593, 608), a result consistent with earlier reports from tively large, thus selection has a considerable impact.
Europe (572). However, differences in the level of protec- Selection for resistance is at least neutral for production
tion conferred by CVI988 from different sources have traits or is correlated positively with production traits
been documented (214, 608). Recent studies demon- (13). Sufficient heterogeneity exists in single‐sire families
strate that a recombinant vaccines with the deletion of to warrant selection for resistance in commercial chick-
both copies of the oncogene meq provides even better ens, which is still the case in recent commercial genetic
protection than the most protective CVI988 strain (115). stocks (184).
The propensity of MDVs to evolve to greater virulence
is critical to the strategic use of vaccines for MD control Selection Methods
(210). Vaccination itself no doubt contributes to this vir- Selection programs for resistance historically have been
ulence increase, which, in turn, tends to make earlier based on progeny testing or family selection (133) or
vaccines obsolete. Kreager (320) noted that the useful life reproduction from survivors of exposed nonvaccinated
of a MD vaccine has been about 10 years under current breeding flocks through mass selection (355). Bacon and
management conditions. Although this is perhaps an Witter (21) found that resistance may better be deter-
overstatement, the implications are serious. Since mined in vaccinated stocks. Under certain conditions
CVI988 was introduced in the United States, some evi- acquisition of resistance can be obtained within four to
dence already suggests that contemporary strains have six generations (133, 355). Family selection may be more
increased their virulence in CVI988‐vaccinated chickens appropriate than mass selection for commercial breeders
(599). In a recent study, Dunn et al. demonstrated that in in order to avoid high loss of genetic material on initial
those farms where vv+MDV were isolated in late 1990s challenge exposure (20). Resistance has been considered
Chapter 15  Neoplastic Diseases 585

dominant, although this varies to some extent (255). In concept to select the most appropriate vaccine based on
most cases, resistance of crosses has been intermediate the predominant B‐haplotypes in a particular strain (23).
to that of the parent strains (55). In practice, this issue has been either ignored or
In addition to selection based on challenge with MDV, addressed through the use of vaccines containing multi-
blood typing for MHC can be used based on the close ple serotypes. Recently, vaccine efficacy has been shown
relationship between MD resistance and certain alleles to be influenced by non‐MCH genetic variation (116).
of the B‐F region of the MHC, especially B21 (61, 71). In light of the selection tools available, the absence of
However, the value of selection for MHC‐associated negative correlations, and the major benefits to be
markers may vary considerably among commercial lines derived, it is not surprising that breeders place a high
and crosses (55, 247). priority on this approach. Although selection for B‐hap-
Several non‐MHC genes may also be involved in resist- lotype has been practiced with variable success and has
ance. For example, line 6 and line 7 chickens differ mark- proven to be complex, especially in meat strains (366),
edly in MD susceptibility, but both lines are both breeders acknowledge the value of improved genetic
homozygous for the MHC B2 allele (143). Because non‐ resistance to offset virulence increase by viral strains and
MHC effects were considered more important than the limitations of current vaccines (319, 624).
MHC effects in several commercial lines (234) the need
to identify genetic markers associated with MD resist-
Management Procedures
ance became priority in all genomic studies. Various
strategies have been used to identify genetic markers of Strict biosecurity practices to limit the extent of early
MD resistance including identification and mapping of MDV exposure, although impractical as a primary con-
map quantitative trait loci (QTL), RNA expression trol procedure, are a crucial and cost‐effective adjunct to
­profiling, expression quantitative trait loci, and allele‐ vaccination. Marek’s disease control is compromised
specific expression (ASE) screens (reviewed in 120). because modern poultry management decisions are
Genome‐wide QTL scans with microsatellite markers often linked to cost analyses. As a consequence replace-
have identified 14 or more putative QTL associated with ment flocks of different ages are placed in close proxim-
MD resistance (70, 569, 632). To complement the QTL ity to each other, vaccine doses are reduced and/or litter
scans, gene expression profiling using microarray from a previous broiler flocks is used (489). The failure
­technology has been integrated. Gene profiling has been to prevent early exposure is perhaps the most important
conducted to identify differentially expressed genes single cause of vaccine failures. Improved hygiene has
between MD‐resistant and MD‐susceptible lines after often appeared to play a key and cost‐effective role in the
MDV challenge (346); among MHC‐congenic lines of elimination of excessive MD losses in vaccinated flocks.
chickens following inoculation with different MD vac- Relevant sanitation principles have been reviewed (423).
cines(375); and in CEF transformed with Meq (341). For SPF flocks, higher standards of biosecurity are
Marek’s disease virus‐chicken protein–protein interac- required and become cost effective. Most SPF operations
tions have been very useful to confirm genes associated use filtered‐air, positive‐pressure houses, which, along
with MD resistance(397) such as growth hormone (GH1) with strict biosecurity measures, successfully can main-
(346), stem cell antigen 2 (SCA2) (347), and MHC class II tain large flocks free of MDV infection. In this case, bios-
β chain (B‐LB)(396). However, purely genetic‐driven ecurity becomes a substitute for vaccination and provides
approaches to identify high‐confidence candidate genes a practical demonstration that MDV infection can be
have not been as successful as first thought. A combina- prevented, at least under specialized conditions.
tion of genetic and functional genomic approaches such
as ASE, epigenetics, and RNA expression profiling seems
to be more useful in identifying candidate genes (121, Nononcogenic Avian Herpesviruses
308, 350, 358, 436, 634, 640).
As mentioned earlier, two additional groups of nononco-
Applications to Control genic herpesviruses, MDV‐2 (Gallid herpesvirus 3) and
The knowledge that genetically resistant chickens are HVT (Meleagrid herpesvirus 1), isolated from chickens
protected by vaccination to a greater extent than more (50, 125) and turkeys (307, 613), respectively, are consid-
susceptible strains (542) has fueled interest by commer- ered part of the Mardivirus genus. The classification of
cial breeders to emphasize MD resistance in selection these two viruses as two distinct serotypes, originally
programs. However, synergy between host genetics and based on the recognition of common and distinct anti-
vaccines is complex. Some resistant B‐haplotypes were genic epitopes (576, 577), has been justified from the
demonstrated only by challenge of previously vaccinated data on the complete sequence for serotype 2 strains
chickens (21). However, the relative efficacy of MD vac- HPRS‐24 and SB‐1 (282, 540), and serotype 3 FC126
cines is also influenced by B‐haplotype, leading to the virus strains (11, 316).
586 Section II  Viral Diseases

Interest in MDV‐2 and HVT derives mainly from their activity was stimulated through at least 8 week PI (517).
use as live vaccines against MD. However, both viruses Herpesvirus of turkey can be recovered from infected
occur in nature independent of vaccination, and it seems chickens for long periods, and antibodies persist for life
appropriate to also consider some aspects of their epizoo- (451, 617). The virus is apparently nononcogenic in tur-
tiology and pathogenesis in their natural or alternate avian keys (588), but the possibility of fertility problems in
hosts. Reviews providing additional details on pathogen- HVT‐infected toms has been raised (560). The virus
esis of these infections may be consulted (88, 90). generally causes no clinical disease in intact or immuno-
suppressed chickens (518, 613). However transient B‐
lymphocyte dysfunction (203), atrophy of the bursa and
Turkey Herpesvirus
thymus with high doses (238), and minor cellular infiltra-
Herpesvirus of turkey is endemic and ubiquitous in tions in nerves (194, 617) have been reported. In con-
domestic turkeys (619) and has also been isolated from trast, up to 19% of S‐line chickens infected with HVT in
wild turkeys (135). In chickens, the virus has also become ovo (ED 8) were reported to develop clinical paralysis
ubiquitous because of its widespread usage as vaccines and gross nerve enlargement due to inflammatory type
(67). Increasingly, HVT is used successfully as recombi- lesions (101). Chickens exposed to HVT at ED 14 or ear-
nant live virus vectored vaccines against a number of lier showed higher incidence of immunological tolerance
­diseases (review in 462). resulting in a persistent HVT viremia (650). A possible
The 160 kb‐long HVT genome encodes nearly 100 role for HVT as a predisposing factor in autoimmune
functional genes that include sets of homologous and disease is suggested by its implied involvement in auto-
unique genes (11, 316), as well as novel microRNAs (579, immune vitiligo in certain strains of chickens (187).
637). The function of HVT genes in relation to their dis- Herpesvirus of turkeys seems to have an adjuvant
tinct properties of high immunogenicity, lack of onco- effect when administered with other vaccines such as
genicity, and ability to produce cell‐free virus are not serotype 2 MDV strains (103, 609) and fowlpox virus
fully understood. The availability of infectious BAC (395). Furthermore, recently it has been shown that
clones of HVT (32) will be helpful in gaining further administration of HVT to chicken embryos at 18 ED has-
insights into HVT gene functions. tens the development of the immune system (221).
In turkeys, HVT spreads rapidly through exposed
flocks by contact exposure as no evidence of vertical
Serotype 2 Marek’s Disease Virus
transmission has been demonstrated (620). Virtually all
individual turkeys become viremic and develop antibod- Apathogenic strains isolated from clinically normal
ies within a few weeks (619). The virus appears to mature chickens (50, 125) subsequently were determined to be a
in the FFE, because cell‐free skin extracts are infectious separate serotype based on antigenic properties (577)
(620) although viral antigen was found only infrequently and genome sequences. The 166 kb‐long MDV‐2 genome
and at low levels in the FFE (194). Herpesvirus of turkey encodes several homologous and unique genes (282,
may be transmitted from turkeys to chickens under 540) as well as novel microRNAs (638). Availability of
experimental conditions (620), although this is consid- infectious BAC clones of SB‐1 virus (437, 529) could
ered extremely rare. Only limited contact spread occurs facilitate functional analysis of MDV‐2 genes.
among chickens, but transmission could not be demon- Serotype 2 viruses are widespread, although not uni-
strated by the airborne route (122). Replication of virus versal in commercial chicken flocks (50). The epizootiol-
in the FFE of infected chickens appears limited and tran- ogy has been complicated by distribution of the virus
sient (122, 446), although increased levels of HVT DNA through a seeder chick program (644) and widespread
were observed in FFE of HVT‐vaccinated chickens after use of MDV‐2 vaccines (103, 616). Other unique features
MDV challenge (278). of this virus group were further elucidated following the
Fabricant et al. (194) studied the early pathogenesis of isolation of the SB‐1 strain (490).
HVT infection in chickens and turkeys. Chickens had no Serotype 2 viruses replicate in the FFE (124) and spread
cytolytic infections in any lymphoid organ. Turkeys readily by contact (490, 618). Following inoculation of
infected with HVT had some viral antigen‐positive cells day‐old chickens, the virus can be first isolated 5–6 days
at 4–14 days PI in the spleen, but no cytolytic infections PI, reaching peak titers at 2–4 weeks, persisting thereafter
in bursa or thymus were seen. In chickens, there was no for long periods (93, 618). A transient splenomegaly was
depression of bursa or thymus size, although a transient induced between 4–12 days PI with SB‐1, but no bursal
splenomegaly was variably present (93, 194). Expression atrophy and only occasional thymic atrophy was seen and
of gB was detected in spleen, with limited levels in thy- cytolytic infection of lymphoid organs was not observed
mus and bursa of Fabricius (264). B cells are rarely (93). In contrast, Lin et  al. (345) found viral antigens in
infected, but latent infection is probably established in spleen and bursal tissues 5–14 days PI, primarily in B cells,
MHC class II‐positive T cells (88). Natural killer cell but no gross or microscopic changes were observed.
Chapter 15  Neoplastic Diseases 587

Calnek (88) considered B cells and macrophages relatively Vaccination with serotype 2 viruses causes a pro-
refractory to infection, and the cells supporting latent nounced enhancement of B‐cell lymphomas in certain
infection lacked MHC class II antigens, thus differing genetic strains of chickens exposed at an early age to sub-
from those in HVT infections. T cells also may not be very group A ALV (24) or REV (12) (see Leukosis/Sarcoma
susceptible because of the poor re‐isolation rates of SB‐1 Group). The ability of serotype 2 virus to enhance LL was
from CD4+ and CD8+ cells of infected chickens (337), attenuated without abrogation of its protective proper-
although MDV‐transformed MSB‐1 cells can be dually ties against MD challenge (594). Avian leukosis virus‐A
infected with MDV‐1 and MDV‐2 viruses (638). SB‐1 is has now been eradicated from most of the chicken lines
not normally considered immunosuppressive (181) susceptible to serotype 2 enhancement and field prob-
although a diminished response to a B lymphocyte‐­ lems due to serotype 2 enhancement of LL are rare (see
specific mitogen and decreased antibody responses to Leukosis/Sarcoma Group). Recently, it has been shown
bovine serum albumen have been reported in chickens that serotype 2 MDV can enhance the development of
vaccinated with SB‐1 strain in combination with HVT. spontaneous ALV‐like bursal lymphomas in ALVA6
However, as it has not been associated with neoplastic transgenic chickens (resistant to infection with sub-
lesions in chickens or embryos, SB‐1 is designated nonon- groups A and E of ALV) (112). Such enhancement is
cogenic rather than apathogenic (490). The absence of independent of age of vaccination (in ovo vs hatch) and
lymphoma was confirmed by other workers (50, 123, 618), might explain some of the spontaneous lymphomas
although there was one report of visceral lymphomas in 2 observed in commercial flocks that are not related to
of 48 chickens inoculated with the HPRS‐24 strain (440). ALV or REV (112).

­Leukosis/Sarcoma Group
Venugopal Nair

Summary of chickens caused by members that belong to the family


Retroviridae (201). Because of the expansion of the lit-
Agent and  Disease.  Avian leukosis sarcoma group of erature on this disease, it is no longer feasible to cite all
pathogens are retroviruses associated with a number of relevant publications that provide the scientific basis for
neoplastic diseases in poultry. These viruses are grouped our current knowledge of the disease. Hence the litera-
into different envelope subgroups and induce diseases ture is cited selectively, with reviews often used instead
such as lymphoid leukosis (LL) and myeloid leukosis of original papers. For more detailed references to the
(ML) that are widespread in many countries causing literature, readers are advised to refer to the chapter in
major economic losses and animal welfare issues. the previous edition (266).
Lymphoid leukosis (LL) has been the most common
Diagnosis.  Major challenge in the clinical diagnosis of form of L/S group of diseases seen in field flocks,
the disease is because of the difficulty in differentiating although myeloid leukosis (ML) has also become preva-
from other avian neoplastic diseases. Flock‐level lent. The neoplasms and their synonyms are listed in
monitoring for the presence of viral antigens, combined Table  15.4. Members of this group of avian viruses are
with serological and molecular diagnostic tests are characterized, as are all members of the Retroviridae, by
important disease diagnosis and eradication. possession of an enzyme reverse transcriptase, which
directs the synthesis of the proviral DNA form of the
Intervention.  Eradication of the pathogen by application RNA virus that forms part of the retroviral life cycle and
of flock monitoring tests to eliminate infected birds at the from which the family name is derived.
pedigree breeding flock level is the best intervention Sections reflecting the host response (Pathology and
strategy. Selection of birds for genetic resistance to Pathogenesis of the Infectious Process) are discussed
infection will also be a useful adjunct in the control strategy. under the pathologic entities without regard for the
properties of the inducing virus(es) other than their
inclusion in the L/S group.
Introduction
Economic Significance
Definition and Synonyms
Infection of chickens with avian leukosis virus (ALV) is
The leukosis/sarcoma (L/S) group of diseases designates the most common L/S virus infection encountered in
a variety of transmissible benign and malignant neoplasms field flocks and is known to be of significant economic
588 Section II  Viral Diseases

Table 15.4  Neoplasms caused by viruses of the leukosis/sarcoma group.

Neoplasm Synonyms

Leukoses
Lymphoid leukosis Big liver disease, lymphatic leukosis, visceral lymphoma, lymphocytoma,
lymphomatosis, visceral lymphomatosis, lymphoid leukosis
Erythroblastosis Leukemia, intravascular lymphoid leukosis, erythroleukosis,
erythromyelosis, erythroblastosis, erythroid leukosis
Myeloblastosis Leukemic myeloid leukosis, leukomyelosis, myelomatosis, myeloblastosis,
granuloblastosis, myeloid leukosis
Myelocytoma(tosis) Myelocytoma, aleukemic myeloid leukosis, leukochloroma, myelomatosis
Connective tissue tumors
Fibroma and fibrosarcoma
Myxoma and myxosarcoma
Histiocytic sarcoma
Chondroma
Osteoma and osteogenic sarcoma
Epithelial tumors
Nephroblastoma Embryonal nephroma, renal adenocarcinoma, adenosarcoma,
nephroblastoma, cystadenoma
Nephroma Papillary cystadenoma, carcinoma of the kidney
Hepatocarcinoma
Adenocarcinoma of the pancreas
Thecoma
Granulosa cell carcinoma
Seminoma Adenocarcinoma of the testis
Squamous cell carcinoma
Endothelial tumors
Hemangioma Hemangiomatosis, endothelioma, hemangioblastomas,
hemangioendotheliomas
Angiosarcoma
Endothelioma
Mesothelioma
Related tumors
Osteopetrosis Marble bone, thick leg disease, sporadic diffuse osteoporostitis,
osteopetrosis gallinarum
Meningioma
Glioma

importance. Economic losses from ALV‐induced dis-


Public Health Significance
eases are attributed to two sources. First, tumor mortal-
ity commonly amounts to around 1–2% of birds, with Recent studies have addressed the relationship between
occasional losses of up to 20% or more. Second, subclini- avian tumor viruses, particularly ALVs, and human
cal infection by ALV, to which most flocks are subject, health. Evidence for the presence of antibodies to ALVs in
produces a depressive effect on a number of important humans usually has been lacking or at best is presumptive
performance traits, including egg production and quality (192). In a serological survey that included 549 human
(152). Economic losses due to ALV tumor mortality and subjects, including groups exposed and not exposed to
reduced productivity are estimated to be in millions of US chickens, significant differences between men and
dollars each year (266). Currently the virus is causing huge women were found for the prevalence of a­ ntibodies to
economic losses to the poultry industry in China (229). ALV but were not related to exposure to chickens (70).
Chapter 15  Neoplastic Diseases 589

Detection of reverse transcriptase activity as a sensitive TM SU


assay for the presence of ALV in human v­ accines derived
from chicken cells was known to be of public health sig-
nificance (332). While the test is of great value in detect- MA
NC
ing contaminating viruses in vaccines (35), no evidence of
antibodies or proviral sequences of ALV was found in the IN RT
vaccines or in the sera of vaccine recipients (187, 352).

History
RNA
CA
The earliest reports of leukotic diseases in fowl are those
of Roloff (334), who described a case of “lymphosar-
comatosis” in 1868, and of Caparini (58), who in 1896
described cases of “fowl leukemia.” Observations of Lipid bilayer PR
osteopetrosis lesions in the bones of chicken recovered Figure 15.18  Schematic diagram of avian leukosis virus particle.
from ancient Roman burial sites were probably associ- The viral envelope is a lipid bilayer in which the gp37
ated with avian leukosis (42). transmembrane (TM) and the gp85 surface (SU) proteins, encoded
Viral oncology was initiated as a discipline from the work by the env gene, are inserted. Internal components encoded by
with the demonstration of transmission of erythroleukemia the gag/pro gene are p19 matrix (MA) protein, p27 capsid (CA)
protein, p12 nucleocapsid (NC) protein, and p15 protease (PR). The
and myelogenous leukemia by inoculating cell‐free filtrates pol gene encodes the reverse transcriptase (RT) and p32 integrase
(116). Peyton Rous received the Nobel Prize in 1966 for his (IN). The core of the particle contains two viral RNA strands.
seminal work on transplantable tumor that could be trans-
mitted by cell‐free filtrates (336). Detailed reviews of the
history of avian retrovirus research are available elsewhere
(107, 266, 298, 306, 338, 389–391, 409, 415, 427). RNA VIRUS

R U5 gag/pro pol env U3 R


Etiology 5′ CAP AAA 3′
Controlling Coding sequences Controlling
Classification sequences sequences

As per the latest classification of viruses (1), avian L/S Reverse transcription
& integration
group are placed in the Alpharetrovirus genus of the
family Retroviridae. Members of this family are RNA DNA PROVIRUS
viruses characterized by the possession of the enzyme 5′ LTR 3′ LTR
reverse transcriptase, which is necessary for the forma- U3 R U5 gag/pro pol env U3 R U5Host DNA
tion of a DNA provirus that is integrated in the host
genome during virus replication. Avian leukosis virus is Controlling Coding sequences Controlling
the type species of the genus (Figures 15.18 and 15.19). sequences Transcription sequences
Rous sarcoma virus (RSV) and a number of replication‐
defective, acutely transforming viruses such as MC29
and MH2 that carry oncogenes are also in the genera.

Translation Messenger RNAs Progeny viral


Morphology genomic RNA

Ultrastructure
In thin‐section electron microscopy, avian leukosis/sar- Structural core Reverse Envelope
proteins: transcriptase (RT); proteins:
coma viruses (ALSV) have an inner, centrally located, p19 (MA), p10, integrase (IN) gp85 (SU),
electron‐dense core about 35–45 nm in diameter, an p27 (CA), gp37 (TM)
p12 (NC);
intermediate membrane, and an outer membrane. This p15 protease (PR)
appearance typifies the C‐type retroviral morphology.
The overall diameter of the virus particle is 80–120 nm, Figure 15.19  Key features of the viral RNA and proviral DNA forms
of the genome of avian leukosis virus. CAP, 5’ end structure; AAA,
with an average of 90 nm. Immature virions budding polyadenylation of 3’ end; R, repeat sequence; U5, unique 5’ end
from the cell membrane can be visualized (Figure 15.20). sequence; U3, unique 3’ end sequence; LTR, long terminal repeat.
Characteristic knobbed spikes about 8 nm in diameter For other abbreviations, see Figure 15.18 and the text.
590 Section II  Viral Diseases

(A) (B)

(C) (D)

Figure 15.20  Ultrastructure of leukosis/sarcoma viruses. (A) BAI‐A of avian myeloblastosis virus (AMV), unfixed, and negatively stained
with neutralized phosphotungstic acid. Peripheral fringe about particles is resolved in some places into discrete “knobs.” ×150,000. (B)
Ultrastructure of leukosis/sarcoma virus release. Virus budding at cell membrane of a leukemic myeloblast. Surface of buds and particles
peripheral to outer membrane is irregular and indistinct (pnu, dense prenucleoid). ×215,000. (C) Thin section of BAI‐A of AMV
sedimented from plasma, fixed in osmium tetroxide, and stained with lead subacetate. Inner and outer membranes and granular
character of nucleoid can be seen. Impression of granules might be derived from sectioning of filaments. Some granules appear to be
hollow. ×510,000. (D) Purified BAI‐A of AMV fixed and shadowed with chromium. ×50,000. (Bonar and de Thé).

are present on the surface of the particles and comprise mL for the buoyant density in sucrose is characteristic
the viral envelope glycoproteins. These projections can for C‐type retroviruses (266).
also be seen in thin sections. Significant advances have
recently been made in ultrastructural studies of retrovi-
Chemical Composition
ral capsids (311, 356).
The overall composition of avian myeloblastosis virus
Size and Density (AMV), which has been studied extensively, is 30–35%
By filtration through membranes of graded pore size, lipid and 60–65% protein, of which 5–7% is glycopro-
ultracentrifugation, and electron microscopy, viruses tein, 2.2% is RNA, and small amounts of DNA are pre-
have a diameter of 80–145 nm. The value of 1.15–1.17 g/ sent, apparently of cellular origin (266). Quantitative
Chapter 15  Neoplastic Diseases 591

proteomics analysis of virus‐infected cells using The pol gene encodes the enzyme reverse transcriptase
improved mass spectrometric methods has identified (RT) present in the core. It is a complex consisting of the
a number of host proteins associated with these b subunit (95 kDa) and the a subunit (68 kDa) derived
viruses (225). from it and has RNA‐ and DNA‐dependent polymerase
and DNA–RNA hybrid‐specific ribonuclease H activi-
Viral Nucleic Acids ties. The b subunit also contains the IN domain (inte-
The major class sizes of RNA sediment at 60–70 S, which grase, p32), the enzyme necessary for integration of viral
is the viral genome, and at 4–5 S, most of which is host DNA into the host genome. Recent structural studies on
tRNA, are thought to be accidentally included in the the catalytic core domain of the ALSV integrase sug-
virion. A tRNA is also associated with the 70 S RNA and gested that it can dimerize in more than one state allow-
serves as a primer for the DNA polymerase during tran- ing the flexibility for multifunctionality during different
scription of viral RNA to DNA. Small amounts of 18 and steps of retroviral life cycle (19).
28 S RNA, viral and cellular mRNA, and DNA are also The virion envelope contains two glycoproteins
present. The 60–70 S genomic RNA is a dimer and can encoded by the env gene: SU (surface, gp85), the viral
be split into two subunits of about 34–38 S, which are surface knob‐like structures that determine viral enve-
believed to represent the diploid genome. These subu- lope subgroup specificity of the ALSV; and TM (trans-
nits of genomic RNA are mRNAs, and their genes have membrane, gp37), representing the transmembrane
been mapped for a number of avian retroviruses. structure that attaches the knobs to the envelope. These
The sequence of the structural genes of ALV, from the 5’ two envelope (Env) proteins are linked to form a dimer,
end to the 3’ end of the RNA molecule, is gag/pro‐pol‐env; termed virion glycoprotein (VGP).
these genes encode, respectively, the proteins of the virion Enzymes and other proteins are found in virions and
group‐specific (gs) antigens and protease, RNA‐depend- are considered to be cellular components incorporated
ent DNA polymerase (reverse transcriptase, or RT), and during virus maturation (393). Of practical value is the
envelope glycoproteins (Figure  15.19). The structural presence in AMV obtained from blood of infected chick-
genes are flanked by terminal genomic sequences with ens, or from myeloblast cultures, of adenosine triphos-
gene promoter and enhancer activities, and that, in the phatase derived from the cell membrane and incorporated
DNA provirus, form the long terminal repeat (LTR) into the virus particle during maturation.
regions. The viral genome is about 7.3 kb in size.
Acutely transforming viruses possess additional trans-
Virus Replication
duced oncogene sequences that initiate neoplastic
transformation. Acquisition of a viral oncogene usually As with other retroviruses, replication of ALSV is char-
is accompanied by genetic defects elsewhere in the viral acterized by the formation, under the direction of reverse
genome (see Pathogenicity). Non‐defective RSV has the transcriptase, of a DNA provirus that becomes linearly
genetic composition gag/pro‐pol‐env‐src. The additional integrated into the host cell genome (Figure  15.19).
gene, src, responsible for sarcomatous transformation, Subsequently, the proviral genes are transcribed into
evidently was acquired originally from a normal cellular viral RNAs, which are translated to produce precursor
oncogene, cellular src. The gene cellular src is an exam- and mature proteins that constitute the virion.
ple of a number of host cell genes, termed proto‐onco-
genes or onc genes, concerned with acute transformation Penetration of the Host Cell
(211, 424). Viral and cellular versions of onc genes, and Detailed reviews describing the recent understanding of
of the specific varieties such as src, are distinguished by the early ALV interactions with the host cells are available
the prefixes v‐ and c‐. Specific v‐onc genes, with c‐onc (266, 393). Although adsorption of the virion to the cell
counterparts in normal cells (Table 15.5). membrane is nonspecific, occurring even in cells resist-
ant to infection, penetration of cells is dependent on the
Viral Proteins presence, in the cell membrane, of host gene‐encoded
The nature, location, and synthesis of proteins that con- receptors specific for particular virus envelope subgroups
stitute avian retroviruses have been extensively studied and on fusion of viral and cell membranes. The receptor
(394) (Figures 15.18 and 15.19). The virion core contains for subgroup A ALV, designated TVA, is related to the
five non‐glycosylated proteins encoded by the gag/pro human low‐density lipoprotein receptor (167, 244).
gene: MA (matrix, p19); p10; CA (capsid, p27), which is Decreased susceptibility subgroup A ASLV in vitro and in
the major gs antigen (Gag) in the core shell; NC (nucle- vivo from intronic deletions in close‐bred line of domes-
ocapsid, p12), involved in RNA processing and packag- tic chickens resulting in inefficient splicing of the tva
ing; and PR (protease, p15), involved in cleavage of mRNA has been reported (63, 328). The receptors for
protein precursors. Other minor polypeptides have been ALV subgroups B, D, and E, designated TVBs3 and TVBs1,
reported. resemble a receptor for cytokines of the tumor necrosis
592 Section II  Viral Diseases

Table 15.5  Acutely transforming avian sarcoma and leukemia viruses classified according to viral oncogene.

Oncogene(s) Oncogene Predominant


Virus strain carried product neoplasm(s) Cells transformed in vitro

RSV, B77, S1, S2 src Nr ptk Sarcoma Fibroblast


FuSV, UR1, PCR II, PCR IV fps Nr ptk Sarcoma Fibroblast
Y73, ESV yes Nr ptk Sarcoma Fibroblast
UR2 ros R ptk Sarcoma Fibroblast
RPL30 eyk R ptk Sarcoma Fibroblast
ASV‐17 jun Tf Sarcoma Fibroblast
ASV‐31 qin Tf Sarcoma Fibroblast
AS42 maf Tf Sarcoma Fibroblast
ASV‐1 crk Ap Sarcoma Fibroblast
AEV‐ES4, erbA, erbB Tf, R ptk Erythroblastosis, Erythroblast, fibroblast
sarcoma
AEV‐R erbA, erbB Tf, R ptk Erythroblastosis Erythroblast
AEV‐H erbB R ptk Erythroblastosis, Erythroblast, fibroblast
sarcoma
S13 sea R ptk Erythroblastosis, Erythroblast, fibroblast
sarcoma
E26 myb, ets Tf Myeloblastosis, Myeloblast, erythroblast
erythroblastosis
AMV myb Tf Myeloblastosis Myeloblast
MC29 myc Tf Myelocytoma, Immature macrophage, fibroblast
endothelioma
CMII myc Tf Myelocytoma Immature macrophage, fibroblast
966 ALV‐J myc Tf Myelocytoma Immature macrophage
OK10 myc Tf Endothelioma Immature macrophage, fibroblast
MH2 myc, mil Tf, S/tk Endothelioma Immature macrophage, fibroblast
Note: Ap 5 Adaptor protein; Nr ptk 5 Nonreceptor protein tyrosine kinase; R ptk 5 Receptor protein tyrosine kinase; S/tk 5 Serine/threonine
kinase; and Tf 5 Transcription factor.

factor family (2–4, 205, 329). Polymorphisms within the Synthesis and Integration of Viral DNA
TVA and TVB receptors have been reported among Detailed reviews on the synthesis and integration of viral
chicken populations (228, 450). Editing of TVB receptor DNA have been provided elsewhere (71, 117, 166, 222,
sequences in DF1 cells can induce resistance to ALV B 383). Major stages in formation of retroviral DNA are:
infections (218). The receptor for the subgroup C avian (1) synthesis of the first (minus) strand of viral DNA by
sarcoma and leukosis viruses, Tvc, is related to mamma- reverse transcription of viral RNA by reverse tran-
lian butyrophilins, members of the immunoglobulin scriptase, forming an RNA : DNA hybrid; (2) removal of
superfamily (115, 263). The host cell receptor used by the RNA from the hybrid by RNase‐H and formation on the
ALV subgroup J, which has a distinct envelope with lim- template of minus‐strand DNA of second (plus) strands
ited homology to those of other subgroups, has been of viral DNA, giving rise to linear DNA duplexes; and (3)
identified as the chicken Na(+)/H(+) exchanger type 1 migration of linear DNA to the cell nucleus. Linear viral
(chNHE1) protein (59, 61, 138, 279), with the noncon- DNA becomes linearly integrated into the host DNA
served tryptophan 38 residue critical in discriminating under the influence of the enzyme integrase. This inte-
resistant and susceptible avian species (206, 327). While gration can occur at many sites, and infected cells can
Japanese quail was shown to be resistant to ALV‐J, some contain up to 20 copies of viral DNA. Recent studies
of the new world quail species with tryptophan 38 residue have demonstrated the importance of host factors such
are susceptible and could potentially serve as reservoirs as the SSRP1 and Spt16 of the FACT protein complex as
of infection (315). Recent study also showed that genetic a principal cellular binding partner of ALV integrase
resistance to ALV‐J could be engineered in DF‐1 cells by (435). The proviral genes occur in the same order as their
genome editing of the receptor sequence (217). RNA copies occur in the virion, and they are flanked on
Chapter 15  Neoplastic Diseases 593

either side by identical sequences of nucleotides—the range, interference pattern, and neutralization can be
long terminal repeats (LTRs) (Figure 15.19). performed more easily with the appropriate RSV pseudo-
type than with the ALV, because the former can be readily
Transcription quantified in cell culture.
Formation of new virions in the infected cell is the result
of transcription and translation of proviral DNA involv-
Endogenous Leukosis Viruses
ing multiple steps (reviewed in 266, 417). Viral RNA
molecules give rise to mRNA in association with polyri- Avian leukosis viruses that are transmitted as infectious
bosomes, and they also serve as genomic RNA in newly virus particles are termed exogenous viruses. The nor-
formed virions. The mRNA species are translated to mal chicken genome also contains several classes or fam-
form the gag, pol, and env gene‐coded proteins that ilies of avian retrovirus‐like elements that are transmitted
­compose the virion. The env gene product is a precursor genetically and are termed endogenous viruses. Extensive
protein gPr92 (92 kDa) from which the viral envelope literature on earlier studies on these elements can be
proteins SU (gp85) and TM (gp37) are derived. accessed in earlier editions of this book (266). Certain
Translation of env is from a spliced subgenomic RNA. retroelements are believed to represent stages in the evo-
The viral proteins localize at the plasma membrane of lution of retroviruses from cellular movable genetic ele-
the cell, where crescent‐shaped structures develop and ments (transposons); whereas others are thought to be
virions that bud off from the cell may be visualized. degenerate proviral forms of exogenous retroviruses that
have lost the ability to produce infectious virus due to
Defectiveness and Helper Viruses mutations.
A number of avian retroviruses (Table  15.6) have been The genetic sequences of the ev loci are related to sub-
shown to have defective genomes and arise either sponta- group E ALVs and are present as either complete or
neously or as a result of experimental mutagenesis (177, defective genomes in almost all normal chickens
238). They will transform cells but require the presence of (Figure 15.21). The chromosomal locations of a number
a helper leukosis virus to enable them to replicate (e.g., of ev loci have been determined using new sequence
BH‐RSV and AMV lack the env gene, and AEV and MC29 analysis pipelines on the chicken genome (32, 342–344).
lack the pol and env genes). Other acutely transforming The phenotypic expressions of these loci vary, depend-
viruses, such as certain strains of RSV, have lost their ing on the viral genes present and on poorly understood
v‐onc gene and ability to transform rapidly. They are control mechanisms (Tables 15.6 and 15.7). More recent
called transformation defective (td) mutants and have an studies have demonstrated the role of endogenous retro-
oncogenic potential similar to that of nondefective ALVs. viruses in generating genomic variations (219) and that
Stocks of rd mutant RSV must by their existence contain the PIWI‐interacting RNAs (piRNAs) can protect germ
helper viruses; these originally were referred to as Rous‐ line from targeting endogenous retroviruses (385). The
associated viruses (RAVs). Infectious RSVs formed in expression of endogenous ev genes is thought to be
these circumstances are called pseudotypes, and their
designation includes the helper virus when this is identi-
fied (e.g., BH‐RSV(RAV‐1) when RAV‐1 strain ALV KB EV
helper virus is used). Use of defective strains of RSV
­enables tailor‐made RSV to be produced with envelope
21 6,10
properties of the helper ALV. Determinations of host
13 7
9.5 1
Table 15.6  Phenotypic expression of representative endogenous 12
avian leukosis viral (ev) genes in normal chicken cells. 6.3 3
2

Phenotype Symbol ev locus

No detectable viral product gs2chf2 1, 4, 5


2 1
Expression of subgroup E envelope gs chf 9
antigen
Coordinate expression of group‐specific gs1chf1 3 15I5 72 151 15I4 15B 63
and envelope antigens
Spontaneous production of subgroup E V‐E1 2 Figure 15.21  Endogenous viral (ev) loci detected in six inbred lines
virus of White Leghorns by restriction fragment polymorphisms
generated after Sac‐1 endonuclease digestion of red blood cell DNA
Source: Adapted from Smith (464). and hybridized to 32P‐labeled RAV‐2 genomic sequences (469).
594 Section II  Viral Diseases

responsible for a dominant form of genetic resistance of sex‐linked gene, K, on the Z chromosome (13), which reg-
chicken cells to infection by subgroup E ALV (ALV E) ulates slow feathering (79). The ev21 gene is expressed as
from a block of virus receptors by envelope protein (239). an infectious endogenous ALV, EV21, in the dam, which is
Endogenous viruses have either beneficial or detri- transmitted congenitally to the progeny, inducing immu-
mental effects, perhaps by their induction of immunity nological tolerance and, consequently, increased suscepti-
or tolerance to tumor virus antigens, depending on when bility to infection by exogenous ALV (13, 175, 363, 365).
they are expressed. Long‐term selection studies showed The biologic functions, if any, of the other endogenous
that only a few of the integrations contributed to the high elements such as endogenous avian retrovirus (EAV),
ALV E expression which seemed to correlate with lower ART‐CH, and CR1 are not fully understood. EAV family
body weights for the females indicating potential linkage are not expressed as infectious virus, but RT activity can
to the loci regulating growth (197). Endogenous viruses be expressed and has been found in live virus vaccines
of the ev family are not essential, because it has been pos- (404, 428). A member of the EAV family, EAV‐HP (also
sible to produce chickens free of ev genes (8). A line of termed ev/J), is believed to be the origin of the env gene of
such chickens has been produced, designated line 0 (81), subgroup J ALV (17). Strongest evidence of the role of
that is of value in research studies and certain diagnostic EAV‐HP in the emergence of ALV‐J by recombination was
tests in which birds or cells free from ev loci are needed. obtained from the identification of an intact chicken EAV‐
Of particular importance is the ev21 locus, which is HP locus showing a uniquely close relationship to the
linked tightly in White Leghorn stock to the dominant ALV‐J prototype clone HPRS‐103 env region (345, 346).

Table 15.7  Phenotypes of endogenous avian leukosis (ev) genes


Susceptibility to Chemical and 
in inbred and commercial lines of White Leghorn chickens. Physical Agents
Details of the susceptibility to chemical agents, condi-
ev Phenotype Line or Sourcea tions of thermal inactivation, pH stability and suscepti-
bility to ultraviolet radiation can be found in the same
1 gs2chf2 Most lines
chapter of the previous editions of this book (266).
1
2 V‐E RPRL72
3 gs1chf1 RPRL63
2 2
Strain Classification
4 gs chf SPAFAS
5 gs2chf2 SPAFAS Antigenicity
Avian leukosis/sarcoma viruses that occur in chickens
6 gs2chf1 RPRL151
have been divided into six envelope subgroups, A, B, C,
7 V‐E1 RPRL15B
D, E, and J, on the basis of differences in their viral enve-
2 2
8 gs chf K18 lope glycoproteins, which determine antigenicity, viral
9 gs2chf1 K18 interference patterns with members of the same and dif-
10 V‐E 1
RPRL 15I4 ferent subgroups, and host range in chicken embryo
11 V‐E1 RPRL 15I4 fibroblasts (CEF) of different phenotypes (266, 306). The
1 other subgroups, F, G, H, and I, represent endogenous
12 V‐E RPRL 151
ALVs occurring in pheasants, partridge, and quail (266).
14 V‐E1 H&N
Occurrence of new ALV subgroup K (ALV‐K) has been
15 (C) None K28 3K16 reported in China based on sequence analysis (107, 224),
16 (D) None K28 3K16 although additional criteria based on the results of
17 2
gs chf 2
RC‐P ­subgroup‐specific neutralization, interference, and host‐
18 V‐E1 RI range analysis will be required for absolute confirmation
1 b of its status as a new subgroup.
19 V‐E (?) RW
Viral interference patterns (Table 15.8) and host range
20 V‐E1(?)b RW
patterns (Tables  15.9 and 15.10) are the most reliable
1
21 V‐E Hyline FP methods for subgroup classification. Antigenicity, as
2 2
Note: Ev13 is associated with the gs chf phenotype, but restriction determined by the production of neutralizing antibodies
fragments have not been characterized. or neutralization by known subgroup‐specific antibod-
a
 Not exclusive to line or source. K, Kimber; R, Reaseheath; H & N, ies, can also be used for strain classification, but is less
Heisdorf and Nelson; for references see Smith (464). dependable. Viruses within a subgroup usually cross‐
b
 The presence of 5 ev loci in Reaseheath line w birds precludes
definitive assignment with the V‐E1 phenotype. Definitive association
neutralize to varying extents, but with the exception of
requires further segregation of ev genes. Hyline FP birds also carry partial cross‐neutralization between subgroup B and D
ev1, ev3, and ev6. viruses, viruses of different subgroups do not.
Chapter 15  Neoplastic Diseases 595

Table 15.8  Interference patterns between avian leukosis virus Molecular Characteristics
(ALV) and Rous sarcoma virus (RSV) of subgroups A–E and J. Sequence analysis of the gp85 encoding sequences of the
env genes of ALVs of subgroups A–E have identified two
Subgroup of challenge RSV hypervariable regions, hr1 and hr2, and three less varia-
Subgroup of ble regions, vr1, vr2, and vr3, in which differences
interfering ALV A B C D E J between the subgroups are present (37, 38, 109). Studies
of recombinants indicated that hr1 and hr2, and to a
A 1 2 2 2 2 2
lesser extent vr3, play the major role in determining
B 2 1 2 1 1 2 receptor tropism (108). However, the exact locations and
C 2 2 1 2 2 2 nature of the differences that determine host range and
D 2 2 2 1 2 2 antigenicity have not yet been identified. The gp85
E 2 2 2 2 1 2 sequence of the env gene of subgroup J ALVs differ more
extensively from those of the other five subgroups, nota-
J 2 2 2 2 2 1
bly at hr1, hr2, vr2, and vr3, and to a lesser extent also
Note: Susceptible avian embryo fibroblast cultures are infected with between these regions (17, 18). Different subgroup J
ALV of each subgroup and challenged several days later with RSV of ­isolates also vary at particular hypervariable regions of
each subgroup. Reduction in RSV foci in infected cultures compared
with uninfected controls is indicative of viral interference. 1,
gp85 (227, 245).
interference; 2, no interference.
Pathogenicity
Table 15.9  Examples of host range of subgroup A–E and J avian Numerous strains of ALSVs exist, many of which were
leukosis/sarcoma viruses in chicken embryo cells of different isolated from naturally occurring or experimentally‐
phenotypes.
induced neoplasms over many years. Many induce a
predominant type of neoplasm and can be named
­
Examples Subgroup of virus accordingly; for example, lymphoid leukosis virus (LLV),
Phenotype (chicken or although ALV is more commonly used than LLV; avian
of cells cell lines) A B C D E J erythroblastosis virus (AEV); avian myeloblastosis virus
(AMV); and avian sarcoma virus (ASV) (Table  15.11).
C/0 15B1 S S S S S S
RPL12 strain of ALV induces lymphoid leukosis, eryth-
C/AE C, alv6 R S S S R S
roblastosis, osteopetrosis, hemangiomas, and sarcomas;
C/A,B,D,E 72 R R S R R S the BAI A strain of AMV induces myeloblastosis,
C/E 0, 15l, BrL S S S S R S lymphoid leukosis, osteopetrosis, nephroblastomas,
­
C/EJ DF‐1/Ja S S S S R R sarcomas, hemangiomas, thecomas, granulosa cell
­
tumors, and epitheliomas (266).
Note: S, susceptible; R, resistant. The cell phenotype designation
denotes chicken (C) cells resistant to (/) the specified subgroup (0, no
Strains of ALSV can also be placed into two major
subgroup; AE, subgroups A and E; etc.). classes in respect of rapidity of induction of tumors:
a
 Cell line, Hunt et al. (265).
1) Acutely transforming viruses. These viruses can
induce neoplastic transformation, in vivo or in vitro,
Table 15.10  Host range of different subgroups of Rous sarcoma
virus (RSV) in embryo fibroblasts of various avian species.
within a few days or weeks. They cause various types
of acute leukemia (leukosis) or solid tumors (usually
sarcomas) (211, 266). The acutely transforming
Subgroup of RSV
viruses are those that carry viral oncogenes in their
genome (Table  15.5). Details of viral oncogenes and
Avian species A B C D E J
the biochemical functions of their products have been
Red jungle fowl S S R R R S reviewed elsewhere (211, 242).
Common pheasant S R R R S R
2) Slowly transforming viruses. These ALVs do not carry
viral oncogenes. They induce tumors by a “promoter
Japanese quail S R R R S R
insertion” or a related mechanism that activates a cel-
Guinea fowl S S S S S R lular oncogene to bring about neoplastic transforma-
Turkey S S S S S S tion, with the development of tumors taking several
Peking duck R R S R R R weeks or months (211, 242).
Goose R R S R R R
Nomenclature
Note: Embryo fibroblast cultures from avian species are challenged
with RSV and susceptibility to RSV focus formation determined. S, A variety of conventions, which reflect the classification
susceptible; R, resistant. Data from Payne et al. (389). methods outlined previously, are used in designating
596 Section II  Viral Diseases

ALSVs; many of these are illustrated in Table  15.11. [BH‐RSV]) or to location (e.g., Prague [PR‐RSV]).
They are given a full and an abbreviated designation Subgroups (e.g., subgroup A) may be designated also:
based on the predominant neoplasm they induce, with PR‐RSV‐A. The general terms avian leukosis (or leuke-
an affix to indicate their origin with an individual (e.g., mia) virus (ALV) and avian sarcoma virus (ASV) are
Rous sarcoma virus [RSV]) or a location (e.g., Regional used widely to ­designate members of the group.
Poultry Research Laboratory isolate 12 [RPL12 of ALV). Helper viruses isolated from stocks of defective viruses
Substrains of RSV are designated according to individu- are named, for example, as Rous‐associated virus (RAV)
als who worked with them (e.g., Bryan’s high‐titer strain or myeloblastosis‐associated virus (MAV), and isolates

Table 15.11  Laboratory strains of avian leukosis/sarcoma viruses of the chicken classified according to predominant neoplasm induced
and virus envelope subgroup.

Virus class according to envelope subgroup

Virus class according to No subgroup


neoplasm A B C D E J (defective virus)a

Lymphoid leukosis RAV‐1 RAV‐2 RAV‐7 RAV‐50 RAV‐60


virus (LLV)
RIF‐1 RAV‐6 RAV‐49 CZAV
MAV‐1 MAV‐2
RPL12
HPRS‐F42
Avian erythroblastosis AEV‐ES4
virus (AEV)
AEV‐R
AEV‐H
Avian myeloblastosis AMV‐BAI‐A
virus (AMV)
Avian sarcoma virus SR‐RSV‐A SR‐RSV‐B B77 SR‐RSV‐D SR‐RSV‐E BH‐RSV
(ASV)
PR‐RSV‐A PR‐RSV‐B PR‐RSV‐C CZ‐RSV PR‐RSV‐E BS‐RSV
EH‐RSV HA‐RSV FuSV
RSV29 PRCII
PRCIV
ESV
Y73
UR1
UR2
S1
S2
Myelocytoma and HPRS‐103 MC29
endothelioma
viruses ADOL‐Hc1 966
MH2
CMII
OK10
RAV‐0
Endogenous virus (EV) EV21
(no neoplasm) ILV‐E
a
 Defective viruses have the envelope subgroup of their helper virus.
Chapter 15  Neoplastic Diseases 597

are numbered (RAV‐1, MAV‐1, etc.). Where a helper


virus is used for replication of a defective virus, this is
indicated. Thus, BH‐RSV grown with RAV‐1 as a helper
is designated BH‐RSV(RAV‐1). Endogenous ALV is
abbreviated EV (e.g., EV21). Strains of ALV that act as
resistance‐inducing factors (see Diagnosis) were desig-
nated RIFs, but this term is now rarely used. Further
details of the origins of the abbreviations (297) are given
in Table 15.11.

Laboratory Host Systems


Chick Inoculation
Rous sarcoma and other sarcoma viruses produce tumors
when injected by the subcutaneous (SC), intramuscular
(IM), or intra‐abdominal (IA) routes and at times by con-
tact with inoculated chickens. Subcutaneous injection
into the wing web or IM injection can be used for sar-
coma virus isolation and propagation (Reviewed in 266).
In susceptible chickens, these may grow rapidly, ulcerate,
and metastasize; in resistant chickens, the sarcomas may
regress. Virus isolates that caused LL also caused eryth-
roblastosis. Osteopetrosis, hemangiomas, and fibrosar-
comas were also observed in chickens of certain strains
and passages (see 266).
Avian myeloblastosis virus can be titrated in suscepti-
ble chicks by IV inoculation at 1–3 days of age. Avian
myeloblastosis virus in chicken plasma can be assayed by Figure 15.22  Pocks induced by BH‐RSV on the chorioallantoic
its adenosine triphosphatase activity, a method useful for membrane (CAM) of a chicken embryo. (Piraino).
routine and large‐scale studies. Osteopetrosis‐inducing
activity of virus strains can be examined by IV or IM
inoculation of day‐old chicks. Guinea fowl are particu- susceptible embryos (20–22). When the HPRS‐103
larly susceptible to osteopetrosis induced by MAV‐2(O) strain of subgroup J ALV was inoculated by IV into 11‐
virus (see 266). day‐old embryos, first death from tumor (myelocytoma)
was not until 9 weeks of age, and median tumor mortality
Embryo Inoculation was at 20 weeks (301).
When RSV and other sarcoma viruses are inoculated
onto the chorioallantoic membrane (CAM) of 11‐day‐ Cell Culture
old susceptible embryos, tumor pocks develop Rous sarcoma virus and other sarcoma viruses induce
(Figure 15.22), which can be counted eight days later and rapid neoplastic transformation of cells when inoculated
are related linearly to virus dose (112). This technique is onto monolayer cultures of chicken embryo fibroblasts
also useful for detecting genetic resistance to infection. (323). The transformed cells proliferate to produce
Avian leukosis viruses have been quantitated by IV within a few days discrete colonies or foci of transformed
inoculation into 11‐day‐old susceptible chicken embryos. cells (Figure  15.23), which under agar can be used for
Depending on the virus, within two weeks of hatching, a quantitative assay of virus (400).
high incidence of neoplasms (mainly erythroblastosis) Most leukosis viruses replicate in fibroblast cultures
can occur, although hemorrhages and solid tumors can without producing any obvious cytopathic effect. Their
develop including fibrosarcomas, endotheliomas, presence can be detected by a variety of tests (see
nephroblastomas, and chondromas. When chicks are Diagnosis). Avian leukosis viruses of subgroups B and D
held for a postinoculation period of 46 days, responses may induce cytopathic plaques that may be used for virus
are higher by 1–2 log10 dilutions than those following assay (163). The cytopathic effect of these two subgroups
chicken inoculation. Most chickens that survive the is explained by their use of the death receptor of the
acute neoplasms develop LL after 100 days PI (312). tumor necrosis factor receptor family (41, 69, 100, 101).
Avian myeloblastosis virus produced a myeloblastosis Acutely transforming ALVs will transform hematopoi-
response within a few weeks when injected by IV into etic cells in vitro (257). Yolk sac and bone marrow cells in
598 Section II  Viral Diseases

(A) (B) (C)

Figure 15.23  Foci induced by Rous sarcoma virus (RSV) in cell culture. (A) Unstained focus of transformed spherical, refractile chicken
embryo cells infected six days previously with Bryan’s standard strain of RSV. ×100. (B) Unstained focus of transformed, polygonal, opaque
Rous sarcoma cells infected six days previously with Bryan’s high‐titer strain. ×100. (C) Unstained focus of transformed round and fusiform
cells infected six days previously with Popken’s preparation of RSV. ×100.

culture are transformed to neoplastic myeloblasts on tumor spectrums of RPL26, RPL27, and RPL28 isolates
infection with AMV (258), and bone marrow cells trans- of ALV (143). Please refer to the chapter in the previous
form to erythroblasts with AEV (164). Transformation of edition for earlier studies in this area (266).
hematopoietic cells by MH2, MV29, and OK10 viruses
was observed by Graf and Beug (165). Acutely trans- Virus Subgroup
forming variants of subgroup J ALV can also transform Usually no relationship has been observed between virus
bone marrow cells and blood monocytes in vitro (66, subgroup and oncogenicity except for endogenous E
302). In vitro transformation of B‐lymphocytes by non- subgroup ALVs, such as RAV‐0, which have little or no
defective ALV has not been reported, and bone marrow oncogenicity (261). However, the low oncogenicity of
cultures were not transformed by nondefective subgroup RAV‐0 is believed to be related to the weak promoter
J ALV (302). activity of the subgroup E LTR and not to the env gene.
The properties of ALSVs in cell culture are described Subgroup J ALV‐induced myelocytomatosis (301), and
in more detail under Diagnosis. the env as well as other elements are thought to be asso-
ciated with the unique oncogenicity (66–68, 234, 235).
Pathogenicity
Virus Dose
As discussed previously (see Strain Classification), High doses of RPL12 ALV mainly induced erythroblasto-
strains of ALVs may produce more than one type of neo- sis; whereas doses close to the endpoint predominantly
plasm, and the oncogenic spectrum of each strain tends induced LL (50). Sarcomas, endotheliomas, and hemor-
to be characteristic but often overlaps with responses to rhages were also more common with high virus doses.
other strains. Viral factors including the origin and dose, Occurrence of osteopetrosis showed no dependency on
and host factors such as route of inoculation, age, geno- dose (144).
type, and sex influence the oncogenic patterns of differ-
ent virus strains. Route of Inoculation
Responses obtained after virus administration by less
Origin of Virus efficient portals of entry into the host apparently reflect
Differences in tumor spectrum may be seen in virus the decreased effective dose. Thus, exposure of suscepti-
strains newly isolated from the field, as exemplified by ble birds by contact with birds inoculated with a high
Chapter 15  Neoplastic Diseases 599

dose of strain RPL12 of ALV resulted in a LL response 133). Molecular and in situ hybridization analysis of the
similar to that expected with 1/1000 of the inoculated bursa from chickens coinfected with ALV and serotype
dose (see 266). Intramuscular inoculation of strain 2  MDV proved that MDV was closely associated with
RPL26 of ALV favored sarcoma induction; whereas IV transformed, but not with nontransformed, bursa cells
inoculation mainly produced erythroblastosis and hem- (148, 240).
orrhages (see 266). These differences may reflect varia- Until recently and before the recognition of subgroup J
tions in amounts of virus that reach the target cells by ALV (299, 308), myelocytomatosis was mainly a sporadic
different routes. disease seen among young and adult birds (321). An over-
all incidence of 27% myelocytomatosis was reported in
Age of Host meat‐type chickens inoculated with strain HPRS‐103 of
In general, resistance of birds to the development of neo- ALV‐J (301). High incidence of ALV‐J tumors have been
plasms of all types increases with age, the rate varying reported in many countries with mortalities up to 1.5% in
with route of inoculation. Resistance increases rapidly excess of normal levels per week in some commercial
between 1 and 21 days of age with oral or nasal adminis- broiler breeder flocks (306). Avian leukosis virus‐J‐
tration but relatively slowly when virus is inoculated induced disease in China is characterized by the occur-
intravenously (48). rence of the disease in meat‐type as well as layer population
including the native breeds (229, 246) and is characterized
Genotype and Sex of Host by a high incidence of hemangiomas (215, 451).
The genetic constitution of the host has a strong influ- Connective tissue tumors, which are often not the
ence on response to ALSVs (see Pathobiology and ­primary cause of death, make up about 20% of nonlym-
Epizootiology). Females are more susceptible to LL than phoid tumors in broilers (57). The incidence of
males. Castration increases the incidence of disease, and connective tissue tumors in chickens is probably less
treatment with testosterone increases resistance of males than 1 in 1,000 (321), but epizootics have occurred. An
and capons (51). These effects are probably a conse- outbreak of histiocytic sarcomas was reported in a flock
quence of hormonal effects influencing regression and, of 600 1‐year‐old hens, during which tumors were found
hence, target cell numbers in the bursa of Fabricius. in 90% of 400 birds examined during a 4‐month period
(310). Low incidence of histiocytic sarcomatosis associ-
ated with ALV‐J has also been reported (5).
Osteopetrosis occurs much less frequently than LL,
Pathobiology and Epidemiology and epizootics occur sporadically in broilers. In all types
of chicken, males are more frequently affected than
Incidence and Distribution
females. Examination of a 1986 outbreak of osteopetrosis
Notwithstanding the eradication programs instituted by revealed ALV sequences (23).
many primary breeding companies, ALV infections still
occur in many flocks. The incidence of subgroup A ALV‐ Incidence of Virus Infection
induced LL, the most common neoplasm observed in Subgroup A ALV is the most common subgroup of L/S
infected flocks, is usually low, in the order of 1 or 2%, viruses isolated from field outbreaks of LL; it is encoun-
although losses of up to 20% can occur. Diseases associ- tered more frequently than subgroup B. In general, fewer
ated with subgroup J continue to be a problem in many studies of the prevalence of ALV in meat lines have been
parts of China (306). made compared with those in egg lines. Antibodies to the
novel subgroup J ALV were found in three of five meat‐
type chicken lines, but not in seven layer lines examined in
Incidence of Disease
the United Kingdom (305). Using virological and serologi-
Earlier reports on the incidence of ALV‐associated dis- cal assays, the incidence of ALV‐J infection in affected
eases can be seen in this chapter in the previous edition broiler breeder flocks was reported to be as high as 87%
of this book (266). Although sporadic cases of ALV‐ (132). Similarly high levels of ALV‐J infection have been
induced neoplasia occur in most flocks, it is only occa- reported in other countries also (139, 226, 229, 246). The
sionally that even the most common neoplasm, LL, incidence of infection with subgroup ALV‐J was also influ-
produces heavy losses. Lymphoid leukosis mortality in enced by other factors such as age at exposure (433, 434).
the Netherlands during 1973–1979 was 2.18% of 11,220 Subgroups A, B, C, and D of ALV have been isolated
white layers and 0.57% of 7,920 brown layers (96, 97), from commercial flocks in Finland; 5 of 10 flocks
although the incidence has been reduced significantly ­surveyed had antibody to all 4 subgroups (348).
since these studies. Serotype 2 MDV was found to Antibodies to subgroups A and B are common among
enhance the development of LL in certain lines of chick- wildfowl and domestic chickens in Kenya and Malaysia,
ens following exposure to ALV after hatch (16, 124, 128, and some evidence exists of antibody to subgroup
600 Section II  Viral Diseases

D viruses in Kenya. Subgroup F viruses have been found (reviewed by 266, 306). Although usually only a small
in ring‐necked and green pheasants, and subgroup ­percentage of chicks are infected vertically, this route of
G  viruses in Ghinghi, silver, and golden pheasants. transmission is important epizootiologically because it
Subgroup H virus has been isolated from Hungarian affords a means of maintaining the infection from one gen-
­partridges and subgroup I virus from Gambel’s quail (see eration to the next. Most chickens become infected by close
Virus Subgroups). Viruses that do not fit within known contact with congenitally infected birds. Although vertical
subgroups have been isolated from Mongolian and transmission is important in the maintenance of the infec-
Swinhoe pheasants, Chinese quail, and chickens. tion, horizontal infection may also be necessary to maintain
However, none were found in Japanese quail, pigeons, a rate of vertical transmission sufficient to prevent the
geese, and Pekin and Muscovy ducks (64, 315). infection from dying out (300). The infection does not
In most vertebrate species, endogenous retroviral spread readily from infected birds to birds in indirect con-
genomes are inherited in a Mendelian fashion and occur tact (in separate pens or cages), probably because of the
at distinct chromosomal loci. DNA sequences related to relatively short life of the virus outside the birds (see
RAV‐0, the endogenous avian retrovirus, occur in the Thermal Inactivation). However, contact exposure at hatch
germ lines of most domestic chickens and several species was shown to be an effective method of spread of ALV‐J
of galliform birds. For example, partridges, true pheas- among broiler breeder chickens (132, 436, 437) and was
ants, grouse, and jungle fowl contain sequences comple- prevented by small group rearing (440).
mentary to RAV‐0; whereas guinea fowl, quail, peafowl, Four classes of ALV infection are recognized in mature
ruffed pheasants, gallo‐pheasants, and turkeys do not chickens: (1) no viremia, no antibody (V−A−‐); (2) no
(145). The structure, function, and regulation of endog- viremia, with antibody (V−A+); (3) with viremia, with
enous retroviruses in the genome of the chicken have antibody (V+A+); and (4) with viremia, no antibody
been reviewed (79, 219, 344, 385). (V+A−) (reviewed by 266, 306). Birds in an infection‐free
flock and genetically resistant birds in a susceptible flock
fall into the category V−A−‐. Genetically susceptible
Natural and Experimental Hosts
birds in an infected flock fall into one of the other three
Chickens are the natural hosts for all viruses of L/S group categories. Most are V−A+, and a minority, usually less
(296); these viruses have not been isolated from other than 10%, are V+A−. Most V+A− hens transmit ALV to a
avian species except pheasants, partridges, and quail (see varying but relatively high proportion of their progeny
Virus Subgroups). Experimentally, however, some mem- (266, 306). A small proportion of V−A+ hens transmit
bers of the L/S group of avian retroviruses have a wide the virus congenitally and do so more intermittently; the
host range and can be adapted to grow in unusual hosts by tendency for congenital transmission of ALV in this cat-
passage in very young animals or through the induction of egory was found to be more frequent in hens with low
immunologic tolerance prior to inoculation of the virus. antibody titer (406). Congenitally infected embryos
Rous sarcoma virus has the widest host range; it will cause develop immunologic tolerance to the virus and after
tumors in chickens, pheasants, guinea fowl, ducks, hatching make up the V+A− class, with high levels of
pigeons, Japanese quail, turkeys, and rock partridges. virus in the blood and tissues and an absence of antibod-
Ducks were shown to be an ideal experimental system for ies. By 22 weeks of age, up to 25% of meat‐type chickens
studying persistence of ALV as the virus appeared to exposed to ALV‐J at hatch were found to be V+A−,
­persist even up to three years with no viremia or neutral- although this could be affected by a number of factors
izing antibodies after embryonic infection (272). However including the virus strain (288, 291).
ducks infected as embryos with subgroup C showed wast- The role of males in the transmission of ALV is at best
ing disease soon after hatching (382, 392, 403). There has equivocal. Infection of the cock apparently does not influ-
been one report of lymphoid leukosis in ostriches (150). ence the rate of congenital infection of progeny (339, 376).
Some strains of RSV induce tumors in mammals (414), The genetics of the host and the strain of ALV influence
including monkeys (207–210). Osteopetrosis can be shedding and congenital transmission after horizontal
­produced in turkeys by inoculation of fresh whole blood infection (90). With electron microscopy, virus budding
from affected chickens (181). Also, turkeys were suscepti- has been seen on all structures of reproductive organs of
ble to ALV‐J infection and tumors induced by the acute cocks except germinal cells (99), indicating that the virus
form of strain HPRS‐103 of ALV‐J (412). does not multiply in germ cells. The cock, therefore, acts
only as a virus carrier and source of contact of venereal
infection to other birds or through semen (226, 366).
Transmission
Congenital infection of embryos is strongly associated
Exogenous ALVs are transmitted in two ways: vertically with shedding by the hen of ALV into egg albumen and
from hen to progeny through the egg and horizontally from with presence of virus in the vagina of hens (304, 375).
bird to bird by direct or indirect contact (Figure  15.24) These traits are also highly correlated with viremia.
Chapter 15  Neoplastic Diseases 601

Shedding of ALV into egg albumen and transmission In flocks infected with subgroup A ALV, only a
to the embryo is a consequence of virus production by minority of ALV‐infected birds develop LL; the others
albumen‐secreting glands of the oviduct. In most hens remain as carriers and shedders. Viremic‐tolerant
congenitally transmitting ALV, the highest titers of (V+A−) birds are reported to be several times more
virus were found in the ampulla of the oviducts, sug- likely to die of LL than those with antibody (V−A+)
gesting that embryo infection is closely related with (339). Incidence of leukosis decreases rapidly if infec-
ALV produced at the oviduct but not with ALV trans- tion by natural routes occurs after the first few weeks
ferred from other parts of the body (405). Electron of age (48); there are well‐established genetic differ-
microscopy studies have revealed a high degree of virus ences in susceptibility to LL development in chickens
replication in the magnum of the oviduct (102). Virus that are equally susceptible to virus infection (88).
budding also occurs in various cell types in the ovary Endogenous ALVs (see Etiology) usually are transmit-
but not in the follicular cells or ovum, and transovarial ted genetically in germ cells of both sexes (Figure 15.24).
infection does not seem to be important (304). Not all Many are genetically defective and incapable of giving
eggs that have ALV in the albumen give rise to infected rise to infectious virions, but some are not and may be
embryos or chicks; in the studies of Spencer et al. (375), expressed in an infectious form in either embryos or
Payne et  al. (304), and Tsukamoto et  al. (406), only hatched birds. In this form, they then are transmitted
about one‐half to one‐eighth of embryos were infected similarly to exogenous viruses, although most chickens
from eggs with virus in the albumen. This intermittent are genetically resistant to such exogenous infection.
congenital transmission may be a consequence of neu- Endogenous viruses have little or no oncogenicity
tralization of virus by antibody in the yolk and of loss but  may influence response of the bird to infection
because of thermal inactivation. by  ­exogenous ALV. Immunodepression induced by

HORIZONTAL

Infectious virus RNA


genome subgroups A & B
or Transient viremia immunity
leukemia rare

VERTICAL

Congenital Genetic
infection transmission

or

Infectious virus RNA Viral DNA genome integrated


genome subgroups A & B in gamete DNA subgroup E

Chronic viremia tolerance Usually latent no viremia


leukemia common no leukemia

Figure 15.24  Horizontal and vertical transmission of exogenous avian leukosis virus (ALV) (e.g., subgroups A and B) and genetic
transmission of endogenous (subgroup E) virus with potential outcomes (115).
602 Section II  Viral Diseases

i­nfectious bursal disease virus increased the rate of MC29 into young chicks, myelocytomas were obtained
­shedding of ALV (135); also strain of virus can influence in 3–11 weeks (253). The incubation period in field
the incidence of tolerantly infected chickens (127). cases is unknown, but most cases are observed in
immature birds. The virus CMII also induces myelocy-
tomas (165). Myelocytomatosis induced by the
Incubation Period
HPRS‐103 strain of ALV, which lacks a viral oncogene,
Members of the L/S group of viruses are multipotent had a long latent period (median time to death was 20
viruses capable of inducing a variety of neoplastic dis- weeks) (301). However, median time to death with the
eases. The incubation period for these diseases is acutely transforming 879‐strain variant of HPRS‐103,
dependent on strain and dose of virus, route of infection, believed to carry a viral oncogene, was nine weeks
age at exposure, and genetic constitution of the host. (302). Field cases of subgroup J‐induced myelocytoma-
Susceptible chicks inoculated as embryos or at 1–14‐ tosis were reported in broiler breeder chickens as young
days‐of‐age with a standard strain of ALV‐RPL12 (50), as four weeks of age (132).
B15, F42 (34), or RAV‐1, developed LL between weeks 14 Most strains of ALV have been found to cause heman-
and 30 weeks of age. It is very seldom that LL cases occur giomas (45, 144). These tumors can be found in birds of
in chickens under 14 weeks. Certain laboratory recombi- various ages. In naturally occurring outbreaks, most
nant viruses have been shown to cause LL within 5–7 mortality from hemangiosarcomas occurred at 6–9
weeks (198), although such short incubation periods are months (54, 55). Induction of lung angiosarcomas by
not found in field outbreaks. subgroup F ALVs is reported (362). After experimental
Another determining factor is whether the virus inoculation of young chicks with field strains of virus
strain lacks or possesses a viral oncogene. For exam- (143), hemangiomas appeared in three weeks to four
ple, diseases such as erythroblastosis, induced by months.
slowly transforming viruses lacking a viral oncogene, In field cases, ALV‐induced renal tumors are rarely
usually develop after a long latent period (67, 146, seen in chickens younger than five weeks; most cases are
211, 422), as in such cases transformation is induced seen in birds between two and six months of age.
by promoter insertion activation of the cellular onco- Nephroblastomas induced by strain BAI‐A may reach an
gene c‐erbB. After IA inoculation of the slowly trans- incidence of 60–85% in birds not dying of myeloblastosis
forming RPL12 strain virus into susceptible day‐old (53). Renal carcinomatous lesions induced by strain
chicks, the incubation period varies from 21–110 MC29 are found as soon as 18 days or as late as 7 weeks
days (50). On IV inoculation of 11‐day‐old embryos, after virus inoculation.
chicks occasionally have been found to have erythro- Osteopetrosis may develop any time after one month
blastosis on hatching. Field strains and viruses pas- following experimental inoculation of day‐old chicks
saged in cell culture induce erythroblastosis after a with strain RPL12‐L29 of ALV (349) or other viruses
longer incubation period (49). Passage from donors (179, 181, 320); it is most commonly seen in birds 8–12
with erythroblastosis greatly shortens the incubation weeks of age. The disease probably has a similar incu-
period (143). bation period in the field. MAV‐2(O) virus will induce
Other strains of virus including F42 (34), ES4, and palpable osteopetrosis 7–10 days after hatching in
strain 13 (28) also produce erythroblastosis. Field cases chicks inoculated at 1 day of age or as 11–12‐day‐old
usually occur in birds older than three months of age. embryos (142).
Viruses such as RPL12 and F42 are non‐defective and Sarcomas may occur any time after inoculation with
slowly transforming; whereas ES4 and R are defective ALVs but are most frequently observed in the first 2–3
and acutely transforming (165). months (52). In field flocks, connective tissue tumors
Strain BAI‐A of ALV predominantly induces myelo- may occur in birds at any age (448). Sarcomas also
blastosis. Virus stocks are defective and contain helper develop readily and are palpable within three days after
viruses of both A and B subgroups (191). After inocula- inoculation of chicks with high doses of acutely trans-
tion of susceptible day‐old chicks with large doses of forming RSV.
virus, changes in the blood can be observed in 10 days,
and birds die a few days thereafter. Mortality continues
Clinical Signs
for about one month, and only a few deaths occur after
this (53, 114). The virus E26 (165) also predominantly Outward signs of the leukotic diseases are mostly non-
induces myeloblastosis. specific. They include inappetence, weakness, diarrhea,
Virus‐induced myelocytomatosis generally has a dehydration, and emaciation. In LL especially, there may
longer incubation period than erythroblastosis and be abdominal enlargement. The comb may be pale,
myeloblastosis induced by the acutely transforming shriveled, or occasionally cyanotic. In erythroblastosis
virus strains, but shorter than LL. On IV injection of and myeloblastosis, hemorrhage from feather follicles
Chapter 15  Neoplastic Diseases 603

also may occur. After clinical signs develop, the course is most tissues and organs of the body (6, 7, 104, 111, 223,
usually rapid, and birds die within a few weeks. Other 333, 381, 402). DiStefano and Dougherty (103, 111)
affected birds may die without showing obvious signs. observed virus budding in cells of every type of tissue
In myelocytomatosis, skeletal myelocytomas may examined, except germ cells and neurons.
cause protuberances on the head, thorax, and shanks. Avian leukosis virus infection in the absence of overt
Myelocytomas may occur in the orbit of the eye, causing disease can adversely affect the productivity of egg‐lay-
hemorrhage and blindness. Hemangiomas may occur in ing chickens. Compared with nonshedders, hens that
the skin, appearing as “blood blisters,” and these may shed virus produced 20–35 fewer eggs per hen housed to
rupture causing hemorrhage (136, 196). Renal tumors 497 days of age; matured later sexually (i.e., age at first
may cause paralysis due to pressure on the sciatic nerve. egg); and produced smaller eggs (374), at a lower rate,
Sarcomas and other connective tissue tumors may be and with thinner shells. Mortality from causes other than
seen in the skin and musculature. When advanced, these neoplasms was 5–15% higher, fertility was 2.4% lower,
various other tumors may be accompanied by the non- and hatchability was 12.4% lower in shedders than non-
specific signs given previously. Benign tumors may shedders (154). In this study, shedding refers to the
­follow a long course, malignant tumors a rapid one. transfer of ALV to egg albumen and nonshedders, as well
In osteopetrosis, the long bones of the limbs are com- as shedders, would likely be ALV‐infected; shedders are
monly affected. Uniform or irregular thickening of the mostly viremic birds; whereas nonshedders are immune
diaphyseal or metaphyseal regions can be detected by with ALV antibody. Avian leukosis virus infection has
inspection or palpation. The affected areas are often similar effects on broiler breeders and causes a consist-
unusually warm. Birds with advanced lesions have char- ent, although often small, reduction in broiler growth
acteristic “bootlike” shanks. Affected birds usually are rate (87, 153). However, these effects are more marked in
stunted and pale and walk with a stilted gait or limp. meat‐type chickens infected with subgroup J ALV (377,
Avian leukosis virus also has been shown to be associ- 378, 380, 381). In broiler breeder flocks affected by mye-
ated with the “so called fowl glioma” (281), associated loid leukosis caused by subgroup J ALV, smaller eggs
with cerebellar hypoplasia and myocarditis (269, 270). were associated with the presence of gs antigen in allan-
toic fluid and virus in the embryo (374). The most criti-
cal time for the immunosuppressive effects of ALV‐J
Pathology
infection in experimental studies was the 3–4 week
Introduction period after infection (420). Other studies on reduced
One or more specific neoplasms induced by ALSVs may productivity in chickens with ALV infections and the
occur in a given flock of chickens, and more than one genetic consequences have been reviewed (152, 189,
type of neoplasm may occur in an individual bird. This is 373). The presence of ALV in semen was not associated
particularly true in flocks infected with subgroup J ALV. with reduced semen production, but some evidence sug-
The presence of a tumor similar to that produced experi- gested an effect on semen quality and fertility (357). The
mentally is only provisional evidence that a bird was physiological bases for these effects have not been
infected with a virus of this group. Firmer evidence is studied.
provided by ALSV detection or isolation, and if appro- A number of other nonneoplastic effects of ALV infection
priate by experimental reproduction of tumors by a virus have been observed, mostly in experimental infections.
isolate. Chickens, turkeys, and jungle fowl exposed when
In this section, the pathology of the different neoplasms young to certain ALVs (RAV‐1, RAV‐60, MAV‐2(O)),
is discussed without regard for virological properties of and ALVs of subgroup B and D develop anemia, hepati-
the inducing agent(s). Only entities that have been repro- tis, immunodepression, and wasting; some may die (82,
duced with ALSVs are described. 371). A myocarditis and chronic circulatory syndrome
was reported in chickens inoculated with RAV‐1 ALV
Nonneoplastic Conditions (158). Intracytoplasmic viral matrix inclusion bodies
The clinical consequences of infection of chickens with have been observed in the myocardium of adult ALV
exogenous ALV vary. Some chickens, principally those infected chickens (156, 267). Chickens inoculated with
with a tolerant viremic infection (arising from congenital RAV‐7 develop neurologic signs including ataxia, leth-
or early neonatal infection, see Immunity), may show a argy, and imbalance resulting from a nonsuppurative
variety of clinical signs, as detailed later in this chapter, meningoencephalomyelitis (429). A persistent infection
including depression of body weight and of other pro- of the central nervous system (CNS), with inflamma-
duction traits. Birds with tolerant viremic infections are tory lesions and clinical signs, followed in ovo infection
also those most likely to develop neoplasia. Ultrastructural with RAV‐1 (120). Fowl glioma‐associated virus also
and virological studies of congenitally or neonatally shows involvement of brain with cerebellar hypoplasia
infected birds have shown the virus to be widespread in (270, 402).
604 Section II  Viral Diseases

Figure 15.25  Nodular lesions in liver and spleen of bird with lymphoma leukosis (LL) inoculated at one day of age with RPL12 virus. Bursa
also has a small tumor.

In MAV‐2(O) infection, anemia occurs due to an aplas-


tic crisis in the bone marrow in which erythrocytes fail to
incorporate iron into hemoglobin and exhibit a decreased
survival time (93). Administration of antiviral antibody
will prevent anemia (320). The immunodepression may
involve atrophy or aplasia of lymphoid organs, hypergam-
maglobulinemia, decreased mitogen‐induced blastogen-
esis, and decreased antibody response (371). The changes
in the immune system are likely a result of cessation of
B‐cell maturation and a block in the development of T‐
suppressor cells, possibly due to interference with the
synthesis of functional interleukin‐2 (179, 214).
In addition to stunting and atrophy of the lymphoid
organs, RAV‐7 caused obesity, high triglyceride and
­cholesterol levels, reduced thyroxine levels (hypothy-
roidism), and increased insulin levels. The frequent
occurrence of stunting may relate to the virus’s suppres-
sion of thyroid function. Stunting of chicks with congen-
ital subgroup J ALV infection was also associated with
hypothyroidism, possibly mediated via effects on the
pituitary (44) or other effects (378).

Lymphoid Leukosis
Gross.  Fully developed LL occurs in chickens of about
four months of age and older. Grossly visible tumors
almost invariably involve the liver (Figures  15.25 and
15.30A), spleen, and bursa of Fabricius (Figures  15.26
and 15.30G). Other organs often grossly involved include
kidney, lung, gonad, heart, bone marrow, and mesentery.
Tumors are soft, smooth, and glistening; a cut surface Figure 15.26  Large tumor of bursa of Fabricius (B) and kidneys
appears grayish to creamy white and seldom has areas of (arrow) in a naturally occurring case of lymphoid leukosis (LL) in
necrosis. Tumor growth may be nodular (Figure 15.25), the adult hen.
Chapter 15  Neoplastic Diseases 605

miliary, diffuse (Figure  15.30A), or a combination of been shown to be remnants of sinusoidal endothelial
these forms. In the nodular form, the lymphoid tumors cells (168). In the bursa, a follicular pattern of tumor
vary from 0.5–5 cm in diameter and may occur singly or growth usually can be seen.
in large numbers. They are usually spherical but may be Tumors consist of aggregates of large lymphoid cells
flattened when they are close to the surface of an organ. (lymphoblasts) that may vary slightly in size but are all at
The miliary form, which is most obvious in the liver, the same early developmental stage. They have a poorly
­consists of numerous small nodules less than 2 mm in defined cytoplasmic membrane, much basophilic cyto-
diameter uniformly distributed throughout the paren- plasm, and a vesicular nucleus in which there are mar-
chyma. In the diffuse form, the organ is uniformly gination and clumping of the chromatin and one or more
enlarged, slightly grayish in color, and usually very conspicuous acidophilic nucleoli (297).
­friable. Occasionally, the liver is firm, fibrous, and almost The cytoplasm of most tumor cells contains a large
gritty. amount of RNA, which stains red with methyl green
pyronin, indicating that the cells are immature and rap-
Microscopic.  All tumors are focal and multicentric in idly dividing see (266). Characteristic features of the cell
origin. Even in organs appearing diffusely involved when can best be seen in wet‐fixed impression smears that
examined grossly, the microscopic pattern is one of have been stained with May–Grunwald–Giemsa, methyl
coalescing foci. As tumor cells proliferate, they displace green pyronin, or other cytological stains. The tumor
and compress cells of the organ rather than infiltrate cells have B‐cell antigen markers and produce and carry
between them (Figure 15.27). Nodules in the liver usually IgM on their surface (77, 307).
are surrounded by a band of fibroblast‐like cells that have
Ultrastructural.  Vacuoles are found infrequently in
lymphoid cells of birds with LL, but some virus particles
have been observed budding from the plasma membranes
of lymphoblasts (for details of references to this work, see
266). Inclusion bodies in enteric smooth muscle and
virions resembling avian retroviruses in the adjacent
intercellular spaces of a bird positive for ALV‐J PCR (171).

Pathogenesis.  Avian leukosis viruses multiply in most


tissues and organs of the body (111). Transitory lymphoid
foci may occur in various tissues and are considered to be
inflammatory in nature (Figure  15.28). The infection
persists longer in bursal lymphocytes than in other
hematopoietic tissues (9, 10), and cells of the bursa of
Fabricius are the target cells that neoplastically transform.
The target cells must be resident in the bursa, because
surgical bursectomy up to five months of age and other
treatments that destroy the bursa of Fabricius will eliminate
the disease. (For details of references, please refer to 266).
Medullary macrophages appear to be the principal bursal
cells for virus replication and may be important in
transmitting infection to the lymphoid cells (157). At a
variable time after infection, which can be as short as four
weeks in experimental studies, a proliferation of
lymphoblasts occurs in one or more lymphoid follicles in
the bursa. These altered bursal follicles are termed
transformed follicles (9, 275, 321), and the change is
regarded as a focal preneoplastic hyperplasia (182, 183)
(Figure 15.29). The transformed follicle is a consequence of
activation of the c‐myc gene by nearby insertion of ALV.
This places the c‐myc gene under the control of the
enhancers of the viral LTR, resulting in over‐expression of
Figure 15.27  Liver tumor from a 20‐week‐old chicken inoculated myc, causing a maturation arrest and proliferation of bursal
at one day of age with RAV‐1 ALV. Note displacement and stem cells, associated with changes in global gene expression
compression of hepatic parenchyma. ×700 (179). profiles and genomic instability (276). Arrest of maturation
606 Section II  Viral Diseases

of the transformed B cells results in interference of the


normal intraclonal switch of immunoglobulin production
from IgM to IgG, hence the surface IgM that characterizes
LL cells. The cells grow within the confines of the bursal
follicle and are not neoplastic. Sometimes, many follicles
are transformed, but the majority of these appear to regress,
and only a few continue to grow to give rise to nodular
tumors in the bursa, which are visible grossly from about 14
weeks of age (76, 275). Progression of the transformed
follicle to the fully neoplastic state requires additional
genetic changes, and other putative oncogenes, Blym‐1
(162, 273) Mtd/Bok (43) and c‐bic (72, 395), have been
implicated. Studies have suggested that the oncogenicity
associated with the non‐coding c‐bic transcript is due to a
novel microRNA designated miR‐155 (396). Oncogenicity
assays demonstrated that bic can cooperate with c‐myc in
lymphomagenesis and erythroleukemogenesis, providing
direct evidence for the involvement of untranslated RNAs
in oncogenesis (397). Genome‐wide analysis of palindrome
formation has shown that myc‐induced genomic instability
from palindrome formation in many sites including the
c‐bic/miR‐155 locus is a major factor triggering the bursal
lymphoma (274). Analysis of the proviral integration sites in
B‐cell lymphomas showed that the telomerase reverse
transcriptase (TERT) promoter/enhancer region was a
common integration site, suggesting that upregulation of
cellular TERT by insertional activation is a factor initiating/
enhancing B‐cell lymphomas (445). Recent studies have
Figure 15.28  Lesions in young chickens induced by leukosis virus. also identified other integration sites that contribute to the
Heart from a 4‐week‐old RIF3‐infected chick showing diffuse development of tumors (195, 196, 309). Evidence also
accumu­lations of lymphoid cells among myocardial fibers ×430. suggests that apoptosis of neoplastic bursal cells is inhibited
(Calnek). by an antagonist of apoptotic cell death, NR‐13, related to
the Bcl‐2 proto‐oncogene (221, 280). Induction of
angiogenic factors from the transformed cells also
contributes to the generation of myc‐induced lymphomas
(40). From about 12 weeks of age, cells in the clonal bursal
tumors metastasize to other organs and tissues and result in
the terminal disease. Metastatic tumors in the viscera
usually have the same DNA fragments as bursal tumors
from the same birds, supporting their clonal origin (83), but
multiple bursal tumors can give rise to polyclonal metastatic
disease (368).
Experimentally, B‐cell lymphomas also have been
induced by c‐myb activation, following embryonic infec-
tion with ALV (198, 313). The tumors were unusual in
that metastatic disease occurred within seven weeks of
infection, and preneoplastic and primary bursal neo-
plasms were not detected. Spontaneous bursal lympho-
mas of unknown etiology have also been observed in
lines of chickens free of exogenous ALV and including
line 0 free of ev loci (91, 92). Reticuloendotheliosis virus
Figure 15.29  Lymphoblastic transformation in single bursal (see Reticuloendotheliosis) can also induce LL associ-
follicle in chicken with lymphoma leukosis (LL). All surrounding
follicles are histologically normal in this and other sections from a ated with c‐myc activation.
16‐day‐old chicken infected with RPL12 virus at hatching. Methyl Transplantable LL tumors can be developed from
green pyronin, ×40. (Dent). ALV‐induced tumors. The RPL12 transplantable tumor,
Chapter 15  Neoplastic Diseases 607

from which the RPL12 strain of ALV was isolated, is a shape and often has pseudopodia. Erythroblasts have cell
well‐known example. Several other new LL transplanta- markers that identify them as members of the erythro-
ble tumors have been described (284). Transplantable LL cytic series.
tumors grow to a palpable size within 5–10 days and Stained blood smears reveal a variable number of
become widely disseminated, inducing rapid mortality. erythroblasts (Figure  15.30D). These vary in maturity
from the early erythroblast, which is the dominant cell,
Erythroblastosis to the various stages of polychrome erythrocytes. The
Gross.  Natural cases of erythroblastosis (erythroid more mature cells often appear early in the course of the
leukosis) usually occur in birds between three and six disease or during remission, if it occurs. The thrombo-
months of age. The liver and kidney are moderately cytic series of cells may be somewhat increased in num-
swollen, and the spleen often is greatly enlarged. The ber and immaturity. Similarly, in most naturally occurring
enlarged organs are usually cherry red to dark mahogany cases, immature cells of the myelocytic series appear in
(Figure 15.30B) and are soft and friable. The marrow is the peripheral circulation. Occasionally, they are as
hyperplastic, semi‐liquid, and red in color. Petechial prominent as the erythroblasts. Cases of mixed erythro-
hemorrhages occur in various organs such as muscles, blastosis and myelocytomatosis may occur.
subcutis, and viscera. Thrombosis, infarction, and
rupture of the liver or spleen may be observed. Edema of Ultrastructural.  Numerous studies have been made of the
the lungs, hydropericardium, and a fibrinous clot on the primitive cells in erythroblastosis induced by different
liver may occur. strains of ALV and RPL12 (for details of these references
With severe anemia, atrophy usually is seen in visceral please refer to 266). Neoplastic erythroblasts are for the
and lymphoid organs, particularly the spleen. most part indistinguishable from corresponding cells in
Changes in the blood reflect those in other organs, the normal bird, except that virus particles may be present
such as liver, spleen, and bone marrow, and depend in extracellular spaces and within vacuoles inside cells. In
largely on the extent of anemia or leukemia. When severe erythroblasts in the circulating blood, as in cell culture,
anemia exists, the blood is watery and light red and clots there is a great increase in membrane activity, with
slowly. In contrast, acute cases may show no grossly vacuolization of the cytoplasm and budding of virus
apparent changes, although usually the blood appears particles from the cell membrane.
dark red with a smoky overcast.
Pathogenesis.  Inoculation of slowly transforming strains
Microscopic.  Examination of the marrow in early cases (i.e., those lacking a viral oncogene) of ALV such as
reveals blood sinusoids filled with rapidly proliferating RPL12 into 11‐day‐old chick embryos induces
erythroblasts that fail to mature. In advanced cases, erythroblastosis from the first week of age (312). When
marrow consists of sheets of homogeneous erythroblasts day‐old chicks are inoculated, the incubation period
with small islands of myelopoietic activity and little or no varies from 21 to more than 100 days (50). Induction of
adipose tissue. With concurrent anemia, the number of erythroblastosis by slowly transforming ALV involves
erythropoietic cells may be reduced. activation of the cellular oncogene c‐erbB by LTR
Alterations in visceral organs are primarily due to insertion (67, 147, 213), and new acutely transforming
hemostasis, resulting in an accumulation of erythro- AEV strains with transduced c‐erbB genes may arise (for
blasts in the blood sinusoids and capillaries (Figure 15.31). references to these, please refer to (266). Whether such
The liver sinusoids, splenic red pulp, bone marrow, and acutely transforming viruses spread naturally and induce
sinusoids of other organs are filled with proliferating more erythroblastosis is not clear.
erythroblasts. Experimentally, acutely transforming AEV strains,
The sinusoids become greatly distended, resulting in such as ES4 and R, cause mortality from erythroblastosis
pressure atrophy of the parenchyma. Although accumu- 7–14 days after inoculation (165). ES4 carries the gene
lations of erythroblasts may be extensive, they always v‐erbA, which blocks erythroid precursor cell differenti-
remain intravascular, unlike those in LL and ation, in addition to the v‐erbB oncogene (for details of
myeloblastosis. these references, please refer to 266). Two subgroup J
Varying degrees of anemia may occur. Sometimes ALV isolates, 1B and 4B, have been shown to be acutely
erythroblastosis occurs, and there may be only severe transforming and to induce erythroblastosis as well as
anemia. Extramedullary erythropoiesis is common. myelocytomatosis and other tumors (411). Their viral
The primary cell involved is the erythroblast. The cell oncogenes have not yet been identified. More recently
has a large round nucleus with very fine chromatin, one ALV‐J‐associated outbreak of erythroblastosis has also
or two nucleoli, and a large amount of cytoplasm that is been reported (421).
basophilic. A perinuclear halo, vacuoles, and occasion- When birds are exposed to an acutely transforming
ally fine granules are present. The cell is irregular in AEV, the first alterations are found in three days as foci of
(A) (B) (C)

(D) (E) (F)

(G) (H)

Figure 15.30  Comparison of leukosis. (A) Lymphoid leukosis (LL): Diffuse form affecting the liver. Lesion is grossly indistinguishable from
those in Marek’s disease (MD); (B) Erythroblastosis: enlarged cherry red liver and spleen, note the fibrinous exudates; (C) Myeloblastosis,
with enlarged gray‐red liver; (D) Erythroblastosis, note the basophilic cytoplasm and perinuclear halo, blood smear, Giemsa stain, ×975;
(E) Myeloblastosis: myeloblasts are slightly smaller than erythroblasts; cytoplasm is not as basophilic, nucleus is less vesicular, and nucleoli
are as not frequent or conspicuous, blood smear, Giemsa stain, ×975; (F) Myelocytomatosis, note myelocytes are packed with acidophilic
granules, section of tumor, Giemsa, ×975 (Beard); (G) LL tumors in the bursa of Fabricius (from the same bird with the liver tumor shown in
(A); (H) Myeloid leukosis tumor on the surface of the skull (Peckham). (For color detail, please see the color section.)
Chapter 15  Neoplastic Diseases 609

Figure 15.32  Myeloblastosis. Distribution of myeloblasts in liver


of bird with myeloblastic leukemia 19 days after inoculation with
BAI‐A virus. ×280. (Langlois).

Figure 15.31  Erythroblastosis. Liver sinuses permeated with


Myeloblasts in leukemic blood smears are large cells
erythroblasts in bird 40 days after inoculation with strain MC29
leukosis virus. ×280. (Beard). with slightly basophilic clear cytoplasm and a large
nucleus containing 1–4 acidophilic nucleoli, which do
not stain prominently (Figure 15.30E). Often, promyelo-
proliferating erythroblasts in bone marrow sinusoids. By cytes and myelocytes are also present; they easily can be
day seven, the primitive cells reach the circulating blood, identified by their specific granulation, which in the early
and some foci of erythroid proliferation are present in forms is primarily basophilic. The disease may result in a
sinusoids of the liver and spleen. Erythroblasts continue secondary anemia, with the presence of polychrome
to accumulate in hepatic sinusoids and elsewhere until erythrocytes and reticulocytes. Such a secondary anemia
death of the host and transplantable erythroblastosis is distinguished easily from the conditions in which
tumors can be developed (316). erythroblastosis and myeloblastosis occur together,
because then blast cells of both cell series are present in
Myeloblastosis the circulating blood.
Gross.  Natural cases of myeloblastosis (myeloblastic
myeloid leukosis) are uncommon and usually occur in Ultrastructural.  In circulating myeloblasts from birds
adult chickens. The liver is greatly enlarged and firm with myeloblastosis induced by BAI‐A AMV, virus
with diffuse, grayish tumor infiltrates, which give a particles are only rarely found and then in small numbers
mottled or granular (“Morocco leather”) appearance in clear vacuoles (28, 105, 172, 173). However, reticular
(Figure  15.30C). The spleen and kidneys are also and phagocytic elements of the spleen and bone marrow
diffusely infiltrated and moderately enlarged. The bone frequently are packed with virus particles. When
marrow is replaced by a solid, yellowish‐gray tumor cell myeloblasts are transferred to cell culture, large numbers
infiltration. of lysosomes appear in the cytoplasm. After some time
A severe leukemia exists, with myeloblasts compris- in cell culture, virus particles can be seen in lysosomes,
ing up to 75% of peripheral blood cells and forming in vacuoles, and budding at the cell membrane. No other
a  thick buffy coat and usually an anemia and changes are observed in these cells.
thrombocytopenia.
Pathogenesis.  The v‐myb gene of AMV is responsible
Microscopic.  Parenchymatous organs, notably the liver, for neoplastic transformation of the target myeloblasts
show a massive intravascular and extravascular accu­ (118). Experimental infection is followed within a few
mulation of myeloblasts with a variable proportion of days by the appearance of multiple foci of proliferating
promyelocytes (Figure 15.32). In the spleen, these tumor myeloblasts in the extrasinusoidal areas of the bone
cells accumulate in the red pulp. In the bone marrow, marrow, followed rapidly by leukemia and infiltration of
myeloblastic activity is confined to extrasinusoidal areas. the liver, spleen, and other organs (for details of these
610 Section II  Viral Diseases

references, please see 266). The v‐myb oncogene acts as


a transcription factor that transforms myelomonocytic
cells by deregulating the expression of specific target
genes as well as through alterations of the nucleosomal
organization (430).

Myelocytomatosis
Gross.  Tumors of myelocytomatosis (myelocytic myeloid
leukosis) are distinctive and can be recognized on
gross  examination with some degree of certainty.
Characteristically, they occur on the surface of bones in
association with the periosteum and near cartilage,
although any tissue or organ can be affected. Myelocytomas
often develop at the costochondral junctions of the ribs,
on the inner sternum, pelvis, and on the cartilaginous
bones of the mandible and nares. Flat bones of the skull
are also commonly affected (Figure 15.30H). Tumors may
also be seen in the oral cavity, trachea, and in and around
the eye (317). The tumors are usually nodular and multiple,
with a soft, friable consistency and of creamy color. In the
disease caused by subgroup J ALV, myelocytomatous
infiltration often causes enlargement of the liver and
spleen and other organs, in addition to skeletal tumors
(432). Myelocytic leukemia may also occur (299).

Microscopic.  Tumors consist of masses of uniform,


usually well‐differentiated, myelocytes. Their nuclei are
large, vesicular, and usually eccentrically located, and a
distinct nucleolus is usually present. The cytoplasm is
usually tightly packed with acidophilic granules, which
Figure 15.33  Myelocytomatosis. Granulated myelocytes in blood
are usually spherical. When imprint preparations of smear from bird 23 days after inoculation with strain MC29
fresh tumors are stained with May–Grunwald–Giemsa, leukosis virus. ×750. (Beard).
granules appear brilliant red (Figure  15.30F). Areas of
less well‐differentiated myelocytes are not uncommon
within the myelocytomas, and areas of undifferentiated Pathogenesis.  Acutely transforming strains of ALV that
cells, which may be stem cells of the myelocyte– induce myelocytomatosis, such as MC29 and CMII,
monocyte series, may also be found. In the liver, carry the v‐myc oncogene (118, 257). Slowly transform­
accumulations of neoplastic myelocytes occur around ing strains of subgroup J ALV that also induce
blood vessels and in the parenchyma. In the spleen, myelocytomatosis, such as HPRS‐103 and ADOL‐Hc1,
tumor cells are present in the red pulp. In the marrow, do not carry an oncogene, but molecular studies of
the extrasinusoidal myelopoietic areas are greatly HPRS‐103‐induced myelocytomatosis indicate that
expanded by uniform neoplastic myelocytes. A detailed c‐myc is activated (66–68). The acutely transforming
description of bone and bone marrow lesions in strain 966 ALV, derived from myelocytoma and induced
myelocytomatosis apparently caused by subgroup J ALV by strain HPRS‐103 of subgroup J ALV, has been shown
is provided by Nakamura et al. (268). to carry v‐myc (66, 302). Studies on HPRS‐103 and 966
Although the naturally occurring disease has been showed that they have a tropism for the myelomonocytic
stated to be usually aleukemic, myelocytomatosis cell lineage, which may relate to their ability to cause
induced by subgroup J ALV frequently is accompanied myelocytomas (6, 7). A recombinant ALV with envelope
by a marked leukemia of myeloid cells. Laboratory strains of subgroup B and LTR of subgroup J (ALV‐B/J) was
of myelocytomatosis‐inducing virus, such as MC29, also isolated from a field outbreak of myeloid leukosis in
cause leukemia (Figure 15.33). commercial layers has been reported (159). However,
inoculation of experimental and commercial strains of
Ultrastructure.  Ultrastructural features of myelocytoma White Leghorn chickens with this recombinant ALV‐B/J
cells vary from those of well‐differentiated myelocytes to resulted in primarily LL, but not myeloid leukosis,
those of undifferentiated, nongranulated myeloid cells (253). suggesting that differences in the genetic makeup of the
Chapter 15  Neoplastic Diseases 611

commercial layers from which ALV‐B/J was originally multiple and may rupture, causing fatal hemorrhage
isolated and lines of chickens used in experimental (372). More recently, many workers have reported the
inoculations studies may be responsible for the incidence hemangiomas in layer chickens infected with
differences in pathogenicity observed (233). ALV‐J in China (215, 287, 454).
The earliest alterations occur in bone marrow in which
there is crowding of intrasinusoidal spaces, principally Microscopic.  The cavernous form is characterized by
by myelocytes, and destruction of sinusoid walls. The greatly distended blood spaces with thin walls composed
spaces may contain two types of cell—the primitive of endothelial cells (Figure 15.35). Capillary hemangiomas
hemocytoblast‐like cell (myeloid stem cell) and the neo- are solid masses in which endothelium may proliferate
plastic myelocyte. The latter appears to arise directly into dense masses (hemangioendothelioma), leaving
from the stem cell, and differentiation is arrested both at mere clefts for blood channels (Figure  15.36); develop
the nongranulated and granulated myelocyte level (253). into a lattice with capillary spaces; or grow into collagen‐
Myelocytes proliferate and soon overgrow the bone mar- supported cords with larger interspersed blood spaces.
row. Tumors form by expansion of marrow growth and Solid and papillary forms have also been described (264).
may crowd through the bone and periosteum.
Extramedullary tumors may also arise by blood‐borne Ultrastructural.  Hemangiomas consisted mainly of
metastasis. undifferentiated mesenchymal cells and had an alveolar
structure (241).
Hemangioma
Gross.  This tumor is found in the skin or in visceral Pathogenesis.  Sequence analysis of an avian
organs in chickens of various ages. They appear as blood‐ hemangioma‐inducing virus isolated from layer hens
filled cystic masses (blood blisters) (Figure  15.34) or revealed unique elements in both env gene and LTR
more solid proliferative, lesions (56). They are often

Figure 15.34  Hemangioma of gizzard serosa of RPL12 virus‐


inoculated bird. Note the dark circumscribed and raised tumor Figure 15.35  Cavernous hemangioendothelioma of mesentery.
nodules (236). (Feldman and Olson).
612 Section II  Viral Diseases

Figure 15.36  Endothelioma in liver of bird inoculated with RPL30


leukosis virus. Occlusion of portal vein by inward‐growing spindle
cells from blood vessel ×250. (Fredrickson).

that were thought probably responsible for its biologic


and pathogenic characteristics (55). This virus was
cytocidal and had an affinity for endothelial cells
(330, 331). Although several sequence changes have
been reported in the genomes of hemangioma‐
inducing ALV‐J strains isolated from layers (287,
443), the precise molecular mechanisms remain
unknown. Recent study has suggested MET gene as a
common integration site in cases of hemangiomas
induced by ALV‐J (196).

Nephroma and Nephroblastoma
Gross.  Two types of renal tumor occur: nephroblastomas Figure 15.37  Nephroblastoma. Bird was inoculated at one day of
(Wilms’ tumor) and adenomas and carcinomas. age with avian myeloblastosis virus BAI‐A strain.
Nephroblastomas vary from small, pinkish‐gray nodules
embedded in the kidney parenchyma to large, yellowish‐ squamous epithelial structures (epithelial pearls),
gray, lobulated masses that replace most of the kidney cartilage, or bone (98, 178, 190). Primary multiplicity of
tissue (Figure 15.37). Tumors may be pedunculated and tumors may occur, but metastases are rare.
connected to the kidney by a thin fibrous vascular stalk. Adenomatous or carcinomatous growths also vary
Large tumors are often cystic and may involve both greatly in microscopic appearance. In tubular adenocar-
kidneys. Adenomas and carcinomas vary in size and cinomas, primitive abnormal glomeruli frequently occur
appearance, similar to nephroblastomas. They are often in large numbers among abnormal tubules. Papillary cyst
multiple and within cysts. adenocarcinomas are frequent. At times, solid carcino-
mas with little evidence of renal tubules develop (30,
Microscopic.  In nephroblastomas, the histologic 253). Rarely is there cartilage and never other mesenchy-
variation between different tumors or areas of the same mal tumor tissues. A trabecular fibrous tissue stroma
tumor is striking. There is usually neoplastic proliferation may separate masses of epithelial tumor tissue.
of both epithelial and mesenchymal elements, although
their proportion and differentiation vary widely. Ultrastructural.  In the epithelial nephronic elements of
Epithelial structures vary from enlarged tubules with nephroblastomas induced by strain BAI‐A of ALV (28),
invaginated epithelium and malformed glomeruli; cytoplasmic aberrant structures occasionally are seen in
through irregular masses of distorted tubules; to groups large or small aggregates. Virus particles bud from cell
of large, irregular, cuboidal, undifferentiated cells with membranes of epithelial cells, fibroblastic elements of
little tubular organization (Figure  15.38). The growth the stroma, and chondrocytes. Sarcomatous elements
may be embedded in a loose mesenchymal or sarcomatous consist of cells similar in morphology to those in other
stroma. There may be islands of keratinizing stratified avian sarcomas. Virus particles have been observed
Chapter 15  Neoplastic Diseases 613

cells form glomeruli, tubules, or keratinized epithelium;


whereas cells of the stroma form sarcomas, cartilage, and
bone. Anaplasia of kidney cells can result in sheets of
large epithelioid cells with almost no tubular organiza-
tion. Malformed and blocked tubules result in cysts.
Nephroblastomas have been induced by BAI‐A strain
AMV (29, 53, 419), MAV‐2(N) (445), MAV‐2‐O (36),
and subgroup J‐related strain 1911 (301). Transplantable
nephroblastomas have been developed (419).
Carcinomatous growths originate only from the epi-
thelial part of the embryonal blastema and not from
mesenchymal elements. Depending on the degree of
anaplasia of epithelial elements, tumors formed may be
adenomas, adenocarcinomas, or solid carcinomas. These
tumors have been induced by MC29, ES4, and MH2
virus strains and by various field isolates. Renal adeno-
mas and carcinomas can be caused by slowly and acutely
transforming subgroup J ALV (for details of these refer-
ences, please see 266).

Fibrosarcoma and Other Connective Tissue Tumors


Gross.  A variety of benign and malignant connective
tissue tumors occur naturally, usually sporadically, in
young and mature chickens, and transmission of many of
these by cell‐free filtrates has been demonstrated. These
tumors include fibromas and fibrosarcomas, myxomas
and myxosarcomas, histiocytic sarcomas, osteomas and
Figure 15.38  Nephroblastoma. Bird was inoculated at one day of
osteosarcomas, and chondromas and chondrosarcomas.
age with cloned preparation of avian myeloblastosis virus BAI‐A The benign tumors grow slowly, are localized, and are
strain. Note primary multiplicity of tumors of two distinct types in noninfiltrative. The malignant counterparts grow more
different areas (arrows) ×20. rapidly, infiltrate surrounding tissue, and may metastasize.
Fibromas arise as firm fibrous lumps attached to the
budding from epithelial cells in cystadenomas and skin, subcutaneous tissues, muscles, and occasionally
adenocarcinomas induced by strain MC29 myelocy­ other organs; fibrosarcomas are of a softer consistency.
tomatosis virus (255). Large accumulations of particles In the skin, they may ulcerate. Myxomas and myxosarco-
in spaces in the cysts and tubules probably were related mas are softer and contain tenacious slimy material.
to a lack of tubule and glomerular drainage. They occur mainly in the skin and muscles. Histiocytic
sarcomas are firm, fleshy tumors occurring mainly in the
Pathogenesis.  A target oncogene for ALV‐induced viscera. Osteomas and osteosarcomas are uncommon
nephroblastomas was not consistently identified (74). and occur as hard tumors that may arise from the perios-
More recently, a new proto‐oncogenes such as nov, (194) teum of any bone. Chondromas and chondrosarcomas
and twist (285) were identified as common integration are rare. They occur where cartilage is present and some-
sites in nephroblastomas (286). times within fibrosarcomas and myxosarcomas.
Nephroblastomas originate from nephrogenic blas- Ganglioneurosarcoma was reported associated with sub-
tema (embryonic nephrons and embryonal rests) (56, group J ALV infection (161).
190). This blastema tissue is present in the metanephros
(functional kidney) at hatching until at least six weeks of Microscopic.  Fibromas in their simplest forms consist of
age and appears as wedge‐shaped foci of immature renal mature fibroblasts interspersed with collagen fibers
tissue particularly beneath the capsule. These epithelial arranged in wavy parallel bands or whorls. Slowly
structures enlarge and become neoplastic. The support- growing tumors are more differentiated and contain
ing stroma of mesenchymal elements also proliferates more collagen and fewer cells than those growing more
and, in turn, may be altered. There is extensive multipli- rapidly. Some fibromas may have edematous areas and
cation of tumor cells (usually convoluted tubules and/or should not be confused with myxomas and
stroma) and varying degrees of differentiation, some myxosarcomas. If necrosis, ulceration, and secondary
abnormal. In the most differentiated form, nephrogenic infection have occurred, various inflammatory and
614 Section II  Viral Diseases

necrotic alterations may be observed in the tumor. Histiocytic sarcomas are derived from cells of the
Inflammatory changes may be so prominent that the monocyte and macrophage lineage, and the cellular con-
tumor may be confused with a granuloma. stituents are highly variable both within a tumor and
Aggressive and destructive growth, their cellular com- between tumors (Figure 15.41). They have been reported
position, and the immaturity of constituent cells associated with infection by subgroup J ALV (5, 169).
(Figure 15.39) characterize fibrosarcomas. Large irregu- Further studies have shown that ALV‐J‐induced histio-
lar and hyperchromic fibroblasts are abundant, and cytic sarcomatosis occurred in persistently viremic but
mitosis is common. Tumors contain less collagen than not immunotolerized meat‐type chickens (289).
fibromas, and this is concentrated in and near irregular
septa that subdivide the tumor. Regions of necrosis often
occur in rapidly growing tumors. Edema is sometimes
present. Multiple undifferentiated pulmonary sarcomas
associated with subgroup J ALV infection have been
reported (170).
Myxomas consist of stellate or spindle‐shaped cells
surrounded by a homogeneous, slightly basophilic,
mucinous matrix. In the malignant form (myxosarcoma),
the mucinous matrix is less abundant, and fibroblasts are
proportionally more numerous and immature than in
myxomas (Figure 15.40). Myxosarcomas associated with
ALV‐A infection have been reported in fancy breed
chickens (431).

Figure 15.40  Myxosarcoma induced by Rous sarcoma virus ×240.


(Helmboldt).

Figure 15.39  Fibrosarcoma in musculature of breast ×120. Figure 15.41  Histiocytic sarcoma of the heart. Note the varied
(Feldman and Olson). character of cellular components ×240. (Helmboldt).
Chapter 15  Neoplastic Diseases 615

The cells may be spindle‐shaped, usually appearing Osteopetrosis


in groups or bundles as in fibrosarcomas; stellate Gross.  The first grossly visible changes occur in the
reticulum‐producing elements; and/or large phago- diaphysis of the tibia and/or tarsometatarsus. Alterations
cytic cells or macrophages. Tumors apparently derived soon are seen in other long bones and bones of the
from stem cells of the myelomonocytic lineage may pelvis, shoulder girdle, and ribs but not the digits.
also be considered to be histiocytic sarcomas. The so‐ Lesions are usually bilaterally symmetric; they first
called endotheliomas induced by MH2 and MC29 may appear as distinct, pale yellow foci against the gray‐
be tumors of this lineage (119). In primary tumors, white, translucent, normal bone. The periosteum is
spindle‐shaped cells usually predominate; whereas in thickened, and the abnormal bone is spongy and at first
metastatic foci, primitive histiocytic forms are more easily cut. The lesion is commonly circumferential and
numerous. advances to the metaphysis, giving the bone a fusiform
Osteomas are structurally similar to bone except that appearance (Figures 15.42 and 15.43). Occasionally, the
much of the inner histologic detail is lacking. They con- lesion remains focal or is eccentric. Severity of the lesion
sist of a homogenous acidophilic matrix of osseomucin varies from a slight exostosis to a massive asymmetric
containing collections of osteoblasts at irregular inter- enlargement with almost complete obliteration of the
vals. Osteosarcomas are usually very cellular infiltrative marrow cavity. In long‐standing cases, the periosteum is
growths that invade and destroy surrounding tissues. not as thickened as it was earlier; when it is removed,
The cells are spindle‐shaped, ovoid, or polyhedral, and the porous irregular surface of the very hard
many are in mitosis. Nuclei are prominent, and cyto- osteopetrotic bone is revealed.
plasm is basophilic. Multinucleated giant cells may be
quite numerous. Although very cellular and rapid grow-
ing, some areas usually have sufficient differentiation for
the production of osseomucin, which is usually sufficient
to identify these tumors.
Chondromas have a typical and unique structure (i.e.,
groups of two or more chondrocytes lying in a matrix of
chondromucin). In chondrosarcomas, considerable cel-
lular variation exists, ranging from the most immature to
the fully mature chondrocyte.

Ultrastructural.  Only the sarcomas produced by RSV


have been examined in detail. The morphology of
fibroblasts, macrophage‐like cells, and mast cells, found
in Rous sarcomas, have been described (28, 172).

Pathogenesis.  Induction of sarcomas and other


connective tissue tumors in the field is likely to be by
activation of a cellular oncogene by a slowly transforming
ALV, occurring up to several months after infection (388).
Avian leukosis virus related to MAV‐1 has been implicated
in a field outbreak of sarcomas in commercial layers;
inoculation of susceptible White Leghorn chickens with
new isolate resulted sarcomas and myelocytomas (448).
Viral oncogenes that have been associated with sar-
coma induction include src, fps, yes, ros, eyk, jun, qin,
maf, crk, sea, and erbB (73, 118, 309, 416) (Table 15.5).
These viral oncogenes reflect the cellular oncogenes
that are activated by insertional mutagenesis and that
may undergo mutation. These cellular oncogenes con-
trol a variety of functions in the cell (their products are
generally growth factors, growth factor receptors, signal
transducers, or DNA transcription factors), and it is
their altered expression that results in the loss of regula- Figure 15.42  Osteopetrosis. A 24‐week‐old chicken, injected with
tion of cell proliferation or differentiation that causes RPL12 at one day of age, with advanced osteopetrotic lesions of
neoplasia. the shanks. (Sanger).
616 Section II  Viral Diseases

(A) (B)

Figure 15.43  Osteopetrosis of tibia in 10‐week‐old chicken. (A) Shorter length of bone is due to reduced growth. Lower tibia is from
control bird of same age. (B) Cross‐section of middle of shaft of bones in A (451).

Early in the disease, the spleen may be slightly enlarged.


Later, severe splenic atrophy occurs as well as premature
bursal and thymic atrophy. Lymphoid leukosis often
occurs in individual birds with osteopetrosis.

Microscopic Lesions.  The periosteum over the lesion is


greatly thickened from an increase in number and size
of basophilic osteoblasts. The number of osteoclasts
per tibia increases, but the density of osteoclasts (i.e.,
the number per unit volume of bone) decreases (354).
Affected bones differ from normal bones in the
following ways. Spongy bone converges centripetally
toward the center of the shaft (Figure  15.44). An
increase occurs in size and irregularity of the haversian
canals, as well as an increase in number and size, and an
alteration in position, of lacunae. Osteocytes are more
numerous, large, and eosinophilic; the new bone is
basophilic and fibrous.
The blood picture is ordinarily aleukemic, and a sec-
ondary anemia often exists. There may be active erythro-
poiesis in remaining bone marrow and sometimes in
focal areas in the liver. Experimentally, viruses that cause
osteopetrosis can induce an aplastic anemia and an
increased corpuscular fragility (179, 320).

Ultrastructural.  Virus particles bud transiently from


osteoblasts and continuously from osteocytes and
Figure 15.44  Osteopetrosis. Cross‐section of humerus from
accumulate in the periosteocytic space. With calcification 8‐week‐old chicken. Six separate osteopetrotic foci are present,
of the bone, the particles become incorporated in the two of which extend from endosteum to periosteum ×18 (451).
bone trabeculae. No virus production is observed from
osteoclasts (141).
ALV (140). Severe cases of osteopetrosis contained 10
Pathogenesis.  Osteopetrosis is a polyclonal disease of times more viral DNA, 30 times more gag precursor
the bone and is thought to be caused by high levels of protein, and 2–3 times more env protein than the infected
virus infection perturbing the growth and differentiation osteoblast cultures. The osteopetrotic lesion is basically
of osteoblasts. Much higher levels of virus infection were proliferative or hypertrophic (57, 349) and may be
found in diseased bones than in cultured osteoblasts neoplastic (39, 354). Lesions of the lymphoid organs and
infected with the Br21 strain of an osteopetrosis‐inducing bone marrow are degenerative or anaplastic (179).
Chapter 15  Neoplastic Diseases 617

The propensity for certain ALVs to induce osteopetrosis tumor growth (335, 359). The presence of cytotoxic lym-
depends on sequences in the gag‐pol‐5’env region of the phocytes against viral envelope antigens has been shown
viral genome (358). Env proteins have also been implicated in birds immunized with ALV or RSV (26, 27, 212), and
in osteopetrosis induction (193). Detection of exogenous cell‐mediated immunity and the major histocompatibil-
ALV sequences most similar to the envelope of ity complex (MHC) complex are clearly implicated in the
myeloblastosis associated virus type 1 (MAV‐1) was regression of Rous sarcomas (350, 351). Viral proteins
identified from the archived bones of an outbreak of expressed on the surface of tumor cells appear to be
osteopetrosis in commercial birds have been reported (23). important targets for the cell‐mediated immunity, and
nonvirion transformation‐specific cell surface antigens
Other Tumors may also be implicated.
Apart from renal tumors, epithelial tumors caused by ALV Chickens that are infected congenitally by ALV do
are uncommon. They mainly have been reported follow- not develop immune responses to the virus. Instead,
ing experimental infections with acutely transforming they become immunologically tolerant to the virus
viruses, although some have occurred in natural and and develop a persistent viremia in the absence of
experimental infections with subgroup J ALV. Strains neutralizing antibodies (248, 340). Inoculating chick-
BAI‐A (29) and HPRS‐103 (301) of ALV have induced the- ens up to two weeks of age with ALV may also induce
comas and granulosa cell tumors of the ovary. A semi- tolerant infection. Early infection with subgroup J
noma in the testis occurred in a bird inoculated with strain ALV is particularly likely to induce a tolerant infection
MH2 (29) and possibly in birds inoculated with subgroup (132, 436, 438). Birds with a tolerant viremic infection
J ALV isolates (308). Adenocarcinomas of the pancreas are more likely to develop neoplasms than are
have been induced in chickens by strains MC29, MH2, immune‐infected birds, because of the greater virus
and HPRS‐103 of ALV (29, 254, 301). The Pts‐56 strain of load in viremics.
osteopetrosis virus produced pancreatic adenomas and Infection by ALV can depress primary and secondary
adenocarcinomas and duodenal papillomas in guinea fowl antibody responses and cell‐mediated immunity (341)
(202–204). Squamous cell carcinomas have been observed to unrelated antigens, although these effects have been
in a few chicks with strains MC29 and MH2 (29). The variable in different studies. Fadly et  al. (129), in a
MC29 and MH2 strains have induced hepatocarcinomas study of congenital infection with an A subgroup ALV,
(29, 216). Other epithelial tumors induced by subgroup J RAV‐1, failed to detect effects on B‐ and T‐cell func-
ALV include cholangioma and ovarian carcinoma (301). tion during the early and late stages of infection, and
Strains MC29 (29) and HPRS‐103 of ALV (301) have they reported no histological damage to the bursa, thy-
been shown to induce mesotheliomas. mus, or spleen. In contrast, subgroup B ALVs have
been reported to induce a marked suppression of the
humoral immune response to several antigens and
Immunity
decreased responsiveness to several mitogens (426).
Active Immunity Evidence that subgroup J ALV is immunosuppressive
Immune responses to oncogenic viruses including ALV appears to be equivocal (136, 230, 231, 379, 380). ALV‐J
have been reviewed (106, 137, 262, 265, 425). Under infection induced a strong immune response at 2‐
natural conditions, most chicks become infected by
­ weeks‐of‐age, but after 4‐weeks‐of‐age, the response
exogenous ALV from penmates or their surroundings decreased quickly suggesting that 3–4 weeks postin-
and, after a transient viremia, develop virus‐neutralizing fection is the critical time at which the ALV‐J virus
antibodies directed against virus envelope antigens that exerts its immunosuppressive effects on the host (420).
rise to a high titer and persist throughout the life of the Nucleotide sequence analysis of consecutive isolates
bird. The virus‐neutralizing antibodies serve to restrict from V+A+ infection profile suggested viral evolution
the amount of virus in the bird, which in turn, will limit to escape the host immune response thereby contrib-
neoplasia, but they generally are considered to have little uting to ALV J persistence (290).
direct influence on tumor growth. After inoculation of
birds with ALV at four weeks of age or older, transient Passive immunity
viremia was detectable at one week and was followed by Serum antibodies, which are mainly in the IgG fraction
antibodies at three weeks and later (237). In a study of (249), are passed on by the hen to her progeny via the egg
birds naturally infected after hatching, antibodies were yolk and provide a passive immunity that lasts 3–4
first detected at 9 weeks of age, with a marked increase in weeks. Passive antibody delays infection by ALV (439),
the proportion with antibodies between 14 and 18 weeks, reduces the incidence of viremia and shedding of ALV
when 80% were positive (340). (122) and reduces the incidence of tumors (46). Level
Antibodies against gs‐antigen may also occur in ALV‐ and persistence of antibody in the chick is related to the
infected birds, but these apparently have no influence on titer of antibody in the dam’s serum.
618 Section II  Viral Diseases

Genetic Resistance (i.e., susceptible to A, B, C, D, E, and J). Recent studies


have shown that precise editing of receptor sequences
Two levels of genetic resistance to leukosis or sarcoma
could be used to induce resistance to infection by ALV
virus‐induced tumors are recognized: cellular resistance
(217, 218).
to virus infection and resistance to tumor development
Chickens with genetic resistance to infection and
(for references to these, please see 266).
tumor induction by different subgroups of ALSVs usu-
Inheritance of cellular resistance to infection is of a
ally fail to develop antibodies (78, 85). Genetic resistance
simple Mendelian type (Table 15.12). Independent auto-
to tumor development has been studied mainly with the
somal loci control responses to infection by ALSVs of
Rous sarcoma (355, 398), regression of which is deter-
subgroups A, B, and C and are designated tva (tumor
mined by a dominant gene, R‐RS‐1, that lies within the
virus A subgroup), tvb, and tvc respectively (79). At each
MHC locus of the chicken and located in the BBL region
tv locus, alleles for susceptibility and resistance exist that
(75, 155, 319, 353). Conserved peptide motifs of the RSV
are designated tvas, tvar; tvbs, tvbr; and tvcs tvcr, respec-
proteins that bind to the MHC have been identified and
tively, and the susceptibility alleles are dominant over the
shown to be immunoprotective against Rous sarcoma
resistance alleles. These genes usually are abbreviated to
growth in chickens with Class I allele B‐F12 (180) and
as, ar, etc. It is probable that multiple alleles occur at each
peptide motifs of the single dominantly expressed class I
locus, encoding different levels of susceptibility (see 232,
molecule explain the MHC‐determined responses to
266, 393).
RSV (418). The MHC (Ea‐B) locus also influences inci-
Mutations in the tvb receptor gene account for the
dence of erythroblastosis and, to a lesser extent, LL (15).
resistance to subgroups B, D, or E infections (25, 184,
Some influence of the lymphocyte antigen Bu‐1 locus on
205, 329). Similarly, intronic deletions that disrupt
Rous sarcoma regression and of the Th‐1 locus on LL is
mRNA splicing of the tva receptor gene was shown to
reported (12). Structural analysis of the MHC alleles in
result in decreased susceptibility to infection by sub-
RSV tumor regression has ruled out the DMA1, DMA2,
group A ALV (328).
BRD2, TAPBP, and BLB2 genes in regression in B6 hap-
Genetic resistance to infection by subgroup J virus has
lotype (386, 387).
not been recognized in chickens, although a number of
other avian species are resistant (306). There is no evi-
dence of the segregation of the ALV‐J cell receptor, Na+/
H+ exchange type I molecule (59), in the chicken popula-
tion (279). However differences between avian species in
Diagnosis
susceptibility to ALV‐J were dependent on the polymor-
Isolation and Identification
phism in this receptor (206, 315, 327).
of Causative Agent
Cellular susceptibility phenotypes associated with
these genes are designated according to a convention Because ALV is widespread among chickens, virus isola-
that recognizes the virus subgroups to which the chicken tion and the demonstration of antigen or antibody have
(C) cell is resistant (/) (e.g., C/AE denotes a cell resistant limited or no value in diagnosing field cases of lympho-
to A and E subgroups but susceptible to B, C, D, and J mas. However, assays for the detection of ALV are very
subgroups); C/0 denotes a cell resistant to no subgroup useful in identification and classification of new isolates,

Table 15.12  Genes controlling cellular susceptibility to leukosis and sarcoma viruses.

Locus Alleles

Virus subgroup Old New Old New Dominant trait

A tva TVA tvas tvar TVA*S TVA*R Susceptibility


s1 r
B and D tvb TBV tvb tvb TVS*S1, S3 TVB*R Susceptibility
C tvc TVC tvcs tvcr TVC*S TVC*R Susceptibility
s r
E tved TVE tve tve TVE*S TVC*R Susceptibility
ie Ie ie Resistance

Note: The locus designation is adapted from Crittenden (117). The new locus designation is that agreed by the
Poultry Committee of the USDA National Animal Genome Research Program, 1994. The allele previously
designated tvbs2 now is considered to be identical to tvbS1. The existence of a tve locus is not settled. The ie locus
is now considered to be an ev locus, with blocking of the subgroup E virus receptor by endogenous virus ENV
glycoprotein expression.
Chapter 15  Neoplastic Diseases 619

safety testing of vaccines, and in testing pathogen‐free test for adenosine triphosphatase activity is specific for
and other breeder flocks for freedom from virus infec- avian myeloblastosis virus. The procedures that are most
tion. Samples most commonly used for detection of ALV widely used have been reviewed extensively in the previ-
include blood, plasma, serum, meconium, cloacal and ous edition of this book as they are not covered here
vaginal swabs, oral washings, egg albumen, embryos, (126, 134, 266). These include the RIF test (337), the
and tumors (265, 266). Virus also can be isolated from COFAL test used to detect the group‐specific antigens of
albumen of newly laid eggs or the 10‐day‐old embryo of ALV (123, 266) and tests based on phenotypic mixing of
eggs laid by hens that are transmitting virus vertically, viruses (80, 266, 282, 322).
from feather pulp, and from semen. All ALSVs are very
thermolabile and can be preserved for long periods only Tests for Viral‐Internal, Group‐Specific (gs) Antigens
at temperatures below −60°C. Thus, materials used for Detection of the major antigen (p27) forms the basis of
biological assays for infectious virus should be collected several diagnostic tests for virus. Highly sensitive
and placed on melting ice or stored at −70°C until ELISA tests for gs antigens are widely used directly for
assayed. In contrast, samples for detection of ALV gs the assay of test material or indirectly using cell cul-
antigens by direct assays can be stored at −20°C; reviewed tures inoculated with test material. These antigens
in (134, 266). may also be detected in cells by FA techniques (200,
Because most strains of ALV produce no visible mor- 294). Using indirect FA tests, monoclonal antibodies to
phologic changes in cell culture, assays for ALV are based ALV‐J proved useful in the detection of ALV‐J infected
on the following: (1) detection of specific proteins or cell cultures (131, 324, 384, 410). A variety of samples
­glycoproteins coded for by one or more of the three can be tested by ELISA for the presence of ALV; how-
major genes of ALV, namely gag, pol, and env genes ever, serum has been shown to be unsuitable for the
(Figure 15.18), or (2) detection of specific proviral DNA detection of exogenous ALV by direct ELISA (303). For
or viral RNA sequences of ALV by the polymerase chain the detection of exogenous ALV, samples are inocu-
reaction (PCR) and reverse transcription (RT)‐PCR, lated on CEFs that are genetically resistant to subgroup
respectively. E ALV. Seven to nine days later, cell lysates are tested
The presence of virus is determined by the detection of for the presence of ALV gs antigen by ELISA (123, 134,
ALV p27 by indirect biologic assays, such as complement 364). Rabbit anti‐p27 antibody, which is used to coat
fixation (CF) for avian leukosis (COFAL), ELISA for ALV, ELISA plates and rabbit anti‐p27 conjugate, as well as
phenotypic mixing, resistance‐inducing factor, and non‐ complete kits for running ELISA for detection of ALV
producer cell activation. The biological assays require gs antigen, are available commercially. An indirect
CEF with specific host range (Table  15.9). Chicken ELISA using a recombinant capsid protein has also
embryo fibroblasts that are resistant to infection with been reported (326).
endogenous ALV (C/E) are desirable to use in tests for
detection and isolation of exogenous ALV. Other cells, Comparison of Tests
such as those resistant to subgroup A (C/A) and resistant In vivo and in vitro cell culture tests for detection or assay
to subgroup J ALV (C/J) (186), can also be used to con- of exogenous ALVs are compared in Table 15.13.
firm the subgroup of isolated ALV. Testing samples on All the in vitro tests require a standard source of
CEFs that are susceptible to all subgroups of ALV (C/O), chicken embryos free from exogenous ALSVs and of
and those that are resistant to subgroup E (C/E) can be known phenotype for use in cell culture. The following
used in differentiating exogenous and endogenous ALV. reagents are also required: for the RIF test, stocks of
If a positive test is obtained from using C/O but not C/E challenge RSV of each subgroup; for the COFAL and
CEFs, the sample is positive for endogenous ALV. Positive ELISA tests, specific antiserum; for the NP test, quanti-
tests using both C/E and C/O indicates the presence of ties of NP cells; and for the PM test, stocks of RSV with
exogenous ALV. Recently, a flow cytometry method endogenous helper, RSV(RAV‐0). Cells obtained from
using a highly specific alloantibody termed R2 has been embryos of unknown genetic origin should not be used
described for detection of endogenous ALV envelope in in RIF, COFAL, and indirect ELISA tests because the
chicken plasma (11, 14). It should be noted that some results may be confused by genetic resistance. Both
tests such as CF and ELISA and possibly non‐producer COFAL and indirect ELISA tests require either pro-
(NP), phenotypic mixing (PM), R(‐)Q cell, and FA can be longed maintenance of culture or several subcultures to
suitable for all leukosis and sarcoma viruses. The resist- propagate the virus sufficiently; therefore, much more
ance‐inducing factor (RIF) test can be performed only on work is involved than in the NP or PM tests.
ALVs that are not rapidly cytopathogenic. Other tests are The subgroup of an infecting ALV can be determined
specific for certain virus strains. Rapid transformation of by any of the tests. In the RIF test, only RSV belonging to
fibroblast cultures is produced only by certain RSV and the same subgroup as the ALV is subjected to interfer-
of hematopoietic cell cultures only by defective ALV. The ence. In COFAL and ELISA tests, genetically resistant
620 Section II  Viral Diseases

Table 15.13  Comparison of methods for assaying exogenous avian leukosis viruses (ALVs).

Response Additional requirements for subgroup Time required


Method Requirements measured determination (days)c

In vivo
Chick inoc 1 day IA LL susceptiblea LL Genetically resistant chickens 270
Chick inoc 1 day IA Erythro susceptibleb Erythro Genetically resistant chickens 63
Embryo inoc 11 days IV Erythro susceptible Erythro Genetically resistant chickens 43
Cell culture RSV pseudotypes, Resistance to Challenge virus of known subgroup 12 + 6
RIF C/E cells formation of
RSV foci in CCd
COFAL Hamster antiserum, Complement Genetically resistant cells 14 + 1
C/E cells fixation
ELISA Enzyme‐linked antisera Color change of Genetically resistant cells 14 + 1
C/E cells substrate
NP NP cells (chicken or RSV foci in CC Genetically resistant cells of RIF test 8+6
quail) with leukosis virus of known
subgroup
PM RSV (RAV‐0), C/O, RSV foci in CC Genetically resistant cells of RIF test 5+6
and C/E cells with leukosis virus of known
subgroup

Note: C/E, cells, genetically resistant to infection with viruses of E subgroup, but susceptible to viruses of other subgroups; C/O, cells
phenotypically susceptible to infection by viruses of all subgroups; COFAL, complement fixation for avian leukosis viruses; CC, cell culture;
ELISA, enzyme‐linked immunosorbent assay; erythro, erythroblastosis; IA, intraabdominal; IV, intravenous; LL, lymphoid leukosis; NP,
nonproducer; RIF, resistance‐inducing factor; RSV, Rous sarcoma virus; RSV (RAV‐0), Rous sarcoma virus with endogenous helper.
a
 Chickens susceptible to LL tumor formation (e.g., line 15I chickens).
b
 Chickens susceptible to virus infection and to development of erythroblastosis (or myeloblastosis).
c
 Approximate number of days necessary to cultivate the virus plus the number of days to indicate the presence of virus.
d
 Cell culture.

cells can be used; thus, an ALV of subgroup A will not infected chickens or in supernatants of myeloblast
produce CF antigens in cells of the C/A phenotype ­cultures (31).
(resistant to subgroup A viruses). In the NP test, geneti- Assays for RT activities have been used for the detec-
cally resistant NP cells can be prepared, and in the PM tion of oncogenic RNA viruses including all ALSVs (399).
test, genetically resistant cells can be used in the mixing Detection of this enzyme, either directly when the cor-
phase. In NP and PM tests, supernatant from the activa- rect template is used (199, 401) or indirectly when the
tion or mixing phase, which contains RSV of the same radioimmunoassay is used (292), is an indication of pres-
subgroup as the ALV, can be placed on genetically resist- ence of virus. Most recently, a highly sensitive PCR‐
ant cells or embryos or used in an interference test with based RT assay has been used to screen human vaccines
an ALV of known subgroup. that are produced in CEF or embryonated eggs for free-
dom from avian retroviruses (188, 236, 404, 413).
Immunohistochemical Tests
Direct (200) and indirect (294) FA tests as well as flow Detection of Viral Nucleic Acids
cytometry (185, 186) have been used to detect viral anti- The PCR is the most common DNA‐based test used for
gen in CEF cultures; flow cytometry has also been shown detection and identification of ALV including subgroup
to be a very useful tool in identifying the subgroup of E viruses (Figure 15.45). Reverse transcriptase‐PCR has
ALV strains contaminating commercial MD vaccines also been used to detect several subgroups of ALV (176,
(24, 121, 361). 454). A specific PCR for ALV subgroup A can be used to
detect proviral DNA and viral RNA in various tissues
Enzyme Assays from ALV‐infected chickens (408). Reverse tran-
Avian myeloblastosis virus has on its surface an enzyme scriptase‐nested PCR (RT‐nested PCR) test that ampli-
(ATPase) that dephosphorylates adenosine triphosphate. fies a fragment of the LTR of exogenous ALV subgroups
This activity can be used as a quantitative assay to deter- A, B, C, D, and J, but not endogenous retroviral sequences
mine the amount of virus present in the plasma of has been described (151). Several primers specific for the
Chapter 15  Neoplastic Diseases 621

(A) the detection of ALV (444). As further modification of


M 1 2 3 4 5 6 7 8 9 10 PCR, loop‐mediated isothermal amplification (LAMP)
method for ALV subgroups have also been developed
(423, 453).

Hematopoietic Transformation
Avian myeloblastosis virus, an acutely transforming
0.3 Kb strain of ALV, harboring an oncogene, can infect and
transform cultures of avian myeloblasts. Assays usually
are based on a quantal response in which individual cul-
tures are scored as positive or negative (21, 256). Focus
(B) assays for myeloblastosis, erythroblastosis, and other
M 1 2 3 4 5 6 7 8 9 10 defective ALVs have been developed (164, 165, 258).
Cultured chicken bone‐marrow cells and blood mono-
cytes are useful in isolation and propagation of acutely
2.3 Kb transforming viruses recovered from cases of myeloid
leukosis induced by strain HPRS‐103 ALV‐J (302).

Transformation of Fibroblasts and Cytopathology


Avian sarcoma viruses transform spindle‐shaped flat
CEFs into spherical and refractile foci (323, 400) that can
be seen microscopically after 4–5 days (Figure  15.23).
(C) Genetically susceptible cultures are inoculated with test
M 1 2 3 4 5 6 7 8 9 10
material. The next day, medium is decanted and is
replaced with an agar overlay (134). Inoculated cultures
should be examined daily for RSV‐induced foci, which
2.3 Kb usually develop within 4–7 days PI.

Serology
Plasma, serum, and egg yolk are suitable samples for the
detection of antibodies to ALSVs.
Figure 15.45  Polymerase chain reaction (PCR) analysis of DNA
isolated from line 0 CEF uninfected and infected with RAV‐1 Tests
(ALV‐A), RAV‐2 (ALV‐B), RAV‐49 (ALV‐C), RAV‐50 (ALV‐D), ADOL‐ Antibody to ALV can be measured by its reaction with
HC‐1 (ALV‐J), and ADOL‐R5‐4 (ALV‐J), and 15B1 cells uninfected RSV or ALV; a virus of one subgroup will not be neutral-
and infected with RAV‐0 (ALV‐E) and EV21 (ALV‐E). (A). PCR analysis ized by antibodies provoked by a virus of a different sub-
using primers specific for ALV‐A‐E. (B). PCR analysis using primers
specific for ALV‐E. (C). PCR analysis using primers specific for ALV‐J
group. Usually, a 1 : 5 dilution of heat‐inactivated (56°C
Lanes: M, 1 kb plus DNA ladder; 1, RAV‐1; 2, RAV‐2; 3, RAV‐49; 4, for 30 minutes) serum is mixed with an equal quantity of
RAV‐50; 5, RAV‐0; 6, EV21; 7, ADOL‐HC‐1; 8, ADOL‐R5‐4; 9 line 0 a standard preparation of RSV of a known pseudotype;
CEF; 10, 15B1 CEF (481, 482). (B. Lupiani). after incubation, the residual virus is quantitated by any
one of many procedures, the cell culture assay being
detection of the most commonly isolated ALVs, particu- most commonly used. A microneutralization test to
larly subgroup A (234), and the new subgroup ALV‐J assay for residual virus can be used for detection of ALV
(360, 369, 370) have been developed. Other primers spe- antibody (134). The test can be conducted in 96‐well
cific for endogenous, subgroup E ALV can also be used microtiter plates, and the neutralization of the virus is
to detect cell culture infected with endogenous ALV‐E, determined by an ELISA on culture fluids (16).
but not those infected with exogenous ALV of subgroups An indirect immunoperoxidase absorbance test (250,
A, B, C, D, and J (134). Recently, a sensitive and specific 251), ELISA tests (252, 367, 405, 407), and flow cytometry
multiplex PCR for detecting subgroups ALV‐A, ALV‐B, (185, 186) have been described for the detection of antibod-
and ALV‐J has been reported (149). Taqman‐based or ies. ELISA kits for the detection of antibodies to ALV
SYBR Green‐based real‐time PCR tests for detection of ­subgroups A and B are available commercially. Also, molec-
ALV have also been used for diagnosis of ALV (94, 325). ularly cloned, baculovirus‐expressed, envelope glycopro-
Use of a proximity ligation technique combined with PCR teins of ALV‐J now are being used in commercial ELISA kits
was used to develop a novel immune‐PCR (Im‐PCR) for specific for the detection of antibody to ALV‐J (220, 410).
622 Section II  Viral Diseases

Serotypes examination. Diagnosis is possible in most instances on


Based on host range, interference spectrum, and viral careful microscopic examination; however, considerable
envelope antigens, viruses of L/S group occurring in experience is necessary. In coming to a decision, history,
chickens are divided into six subgroups A, B, C, D, E, and signs, gross and microscopic lesions, and cytology should
J. Viruses of different subgroups can be distinguished by all be considered. Ordinarily, LL does not occur before
the ability of monovalent antiserums to neutralize them. 14 weeks of age, and most of the mortality occurs
Even though some cross‐neutralization usually exists between 24 and 40 weeks. Marek’s disease, however, may
between viruses belonging to the same subgroup, the occur as early as 4 weeks, and the mortality peak varies
kinetics of neutralization vary, and slopes of curves for from 10–20 weeks. Occasionally, losses continue and
heterologous systems differ from those of homologous may reach a peak after 20 weeks.
systems. No common neutralization antigens are among Nodular tumors of the bursa can often be palpated
the viruses of different subgroups, except for a relation- through the cloaca in birds infected with ALV. Paralysis
ship between subgroups B and D. The diagnosis of infec- associated with gross lesions in autonomic and periph-
tion by serologic means requires that representatives of eral nervous systems and gross lesions of the iris (“gray
all serotypes be employed. Avian leukosis viruses them- eye”) are specific for MD.
selves may be used, but more commonly, RSV pseudo- As stated previously, the bursa of Fabricius plays a cen-
types are employed in the neutralization tests (134). tral role in development of LL. When distinct focal or
nodular lymphoid tumors are present in the bursa, a
diagnosis of LL can be made; however, REV‐induced
Differential Diagnosis
bursal lymphomas must be ruled out. Such tumors are
Lymphoid Leukosis sometimes quite small and may be overlooked. In some
Differential diagnosis of lymphomas in chickens can be birds, MD induces a premature atrophy of the bursa. In
difficult. The two most common lymphoid neoplasms, others, the bursa may be tumorous, in which case the
namely MD and LL are particularly confusing (441). walls and the plica may be thickened from interfollicular
Lymphoid tumors observed in REV‐infected chickens, infiltration with pleomorphic lymphocytes. In contrast,
although only infrequently in cases of use of REV‐con- intrafollicular tumors of the bursa consisting of uniform
taminated vaccines, or under experimental conditions large lymphocytes are usual with LL.
(see Reticuloendotheliosis), may also add to the confu- Microscopic lymphoid infiltration in nerves, cuffing
sion. Lymphoid leukosis cannot be differentiated from around small arterioles in the white matter of the cere-
REV‐induced bursal lymphomas on the basis of pathol- bellum, and the feather follicular pattern of lymphoid cell
ogy, immunohistochemistry, and molecular changes in infiltration in the skin, which are characteristic of MD,
the c‐myc region. Virologic, serologic, or PCR tests may are not seen with LL.
be helpful in establishing infection for one virus and Cytologically, LL tumors generally are composed of a
exclusion for the other. However, such assays are not par- homogeneous population of lymphoblasts (Figure 15.27).
ticularly helpful in the diagnosis of virus‐induced lym- In contrast, tumors of MD usually contain lymphoid
phomas of chickens including LL, as avian oncogenic cells varying in size and maturity from lymphoblasts to
viruses are widespread, and infection in the absence of small lymphocytes, and plasma cells may also be present.
tumor formation is common. Detection of proviral DNA Special stains such as methyl green pyronin are helpful
and integration junctions by PCR assays (67, 160, 314) for cytology. Immature lymphoblasts characteristic of LL
has been shown to be useful for tumor diagnosis. tumors are highly pyroninophilic; whereas the medium
Because LL tumors should contain ALV proviral DNA and small lymphocytes that predominate in tumors of
sequences inserted near the c‐myc gene, differentiation MD do not stain with pyronin.
between LL and REV‐induced bursal lymphomas can be Lymphoid leukosis tumors are composed almost
made by southern blots and hybridization analysis of tumor entirely of B cells and have surface IgM markers; whereas
DNA for clonal insertion of ALV (see previous discussion). 60–90% of MD tumor cells are T cells that lack IgM
Lymphomas in which bursal tumors are lacking or in markers and only about 3–25% are B cells. In addition,
which the latent period is too short for that of LL can be from 0.5–35% of MD tumor cells have a tumor‐associ-
confused primarily with MD; however, under certain cir- ated cell surface antigen (MATSA), which is absent from
cumstances, REV‐induced lymphoma should also be LL tumor cells (113, 277, 278, 318). Recently, Witter et al.
ruled out (see Reticuloendotheliosis). In cases in which (441) introduced a diagnostic strategy for the differential
bursal tumors are lacking, LL and MD can be differenti- diagnosis of viral lymphomas in chickens.
ated only with difficulty, because similar lymphoid Other diseases that may be confused with LL are eryth-
tumors may occur in both diseases in the same visceral roblastosis, myeloblastosis, myelocytomatosis, ­pullorum
organs during the same age period. Visceral lesions of disease, tuberculosis, enterohepatitis, Hjarre’s disease,
these two diseases cannot be distinguished by gross and fatty degeneration of the liver.
Chapter 15  Neoplastic Diseases 623

Erythroblastosis those of LL that specific diagnosis cannot be made with-


Although gross lesions of liver, spleen, and bone marrow out examination of a blood smear. Examination of liver
provide the basis for a presumptive diagnosis, a firm diag- or bone marrow sections is helpful when identity of the
nosis must be based on finding large numbers of erythro- cell type is in doubt. The myeloblast is, on the average,
blasts by microscopic examination of a blood smear and smaller than the erythroblast or lymphoblast; its cyto-
sections or smears of liver and bone marrow. Chickens in plasm is more acidophilic and is polygonal or angular.
early stages of disease or without obvious signs may be The nucleus is less vesicular; the nucleolus, while pre-
missed easily unless microscopic examination is made. sent, is not nearly so frequently seen or conspicuous as in
Erythroblastosis with concurrent anemia is often dif- the other two leukoses. Myeloblasts also have physio-
ficult to differentiate from anemia resulting from non‐ logic markers that identify them as members of the
neoplastic causes. In erythroblastosis, there is usually a ­myeloid series.
defect in maturation of erythroblasts, resulting in the
presence of large numbers of them and very few poly- Myelocytomatosis
chrome erythrocytes. In anemia, the reverse usually The distinctive character and location of tumor (see the
occurs. Extramedullary erythropoiesis and stasis of previous discussion) provide the basis for diagnosis,
erythroblasts in the sinusoids are usually more promi- which can be verified by examination of a stained smear
nent in erythroblastosis than in anemia. or tumor section. Gross tumors must be differentiated
Erythroblastosis can be distinguished from myeloblas- from myeloblastosis, LL, osteopetrosis, and necrotic
tosis on the following grounds. In myeloblastosis, the and/or purulent processes occurring in tuberculosis,
liver is usually pale red and the marrow is whitish; pullorum disease, and mycotic infections. In recent out-
whereas in erythroblastosis, the liver and marrow are breaks of ALV‐J‐induced tumors, myelocytomatosis was
usually cherry red (Figures 15.30B and 15.30C). In mye- diagnosed primarily on the basis of presence of charac-
loblastosis, the cells accumulate intravascularly and teristic microscopic feature of tumor cells (see 266).
extravascularly, whereas in erythroblastosis they are
always intravascular. The erythroblast and myeloblast Hemangioma
may be difficult to distinguish. Erythroblasts have a Hemangiomas on the skin should be differentiated from
basophilic cytoplasm and perinuclear halo; myeloblasts wounds, bleeding feather follicles, and cannibalism.
often have some granules (Figures 15.30D and 15.30E). Those in the visceral organs should be differentiated
Erythroblasts are cells of the erythropoietic system and from hemorrhages and sarcomas.
can be differentiated from cells of the myelopoietic system
on the basis of the presence of certain markers. Thus, Renal Tumors
erythroblasts have erythroid markers including hemo- Renal tumors should be suspected when tumor nodules
globin, chicken erythrocyte‐specific histone H5, and or large masses are found only in the kidney or are
chicken erythrocyte‐specific cell surface antigens detected encountered suspended from the lumbar region.
by immunofluorescence. Myeloblasts and myelocytes have Diagnosis can be verified by microscopic examination.
myeloid markers including adherence and phagocytic Tumors should be differentiated from other causes of
capacity, Fc receptors as determined by rosette formation, kidney enlargement including hematomata, LL, and
macrophage‐ and granulocyte‐specific cell surface antigen accumulation of urates.
as detected by immunofluorescence, and dependence of
colony formation on colony‐stimulating factor (165, 259). Osteopetrosis
Erythroblastosis can be distinguished from LL by the Bone lesions of advanced cases are sufficiently distinc-
nature and distribution of lesions. Microscopically, the tive to present no difficulty in diagnosis. Cross‐ and
cytoplasm of lymphoblasts is somewhat less basophilic ­longitudinal‐sectioning of long bones is helpful in detect-
than that of erythroblasts, and there is also a larger ing slight exostoses and endostoses, particularly in early
nuclear<thin: cytoplasmic ratio than in the latter cells. stages.
Lymphoblasts are more variable in size and shape than
erythroblasts, but they are all at the same primitive
developmental stage. Lymphoblasts tend to have an
ovoid rather than spherical nucleus and a finer, more
Intervention Strategies
delicate‐looking chromatin network. Myelocytomas and
Vaccination
erythroblastosis can be distinguished histologically.
No commercial vaccine is available for the protection of
Myeloblastosis chickens from infection with ALV. However, the idea of
As in erythroblastosis, a tentative diagnosis may be based using vaccines to increase host resistance to ALV infec-
on gross lesions; however, these are often so similar to tion is very attractive (347). In a series of attempts to
624 Section II  Viral Diseases

inactivate ALV by various means, however, Burmester development of ALV‐free flocks, several methods for
(47) demonstrated that ability of these virus preparations selecting dams were used. The dams selected to produce
to induce antibody was destroyed almost concurrently the next generation and hoped to be a virus‐free genera-
with inactivation. Attempts to produce attenuated tion were:
strains of ALV that do not induce disease have also failed
1) immune, non‐virus shedders. Hens with antibody
(283). Results of experimental vaccination with live ALV
were selected on the assumption that they were less
on shedding and congenital transmission of the virus are
likely to shed virus than hens without antibody.
equivocal. Some success has been obtained in attempts
Chicks were hatched from those that did not transmit
to increase the resistance of the host to RSV by immuni-
virus to their embryos, based on tests on at least three
zation with viral or cellular antigens (33, 295).
embryos per hen.
Recombinant ALV‐J gp85 protein vaccine with either a
2) Nonimmune, nonvirus shedders. Hens without anti-
liposomal, a cytosine–phosphate–guanine oligodeoxy-
body were selected on the assumption that they never
nucleotide (CpG‐ODN), or silica nanoparticles adjuvant
been infected and were less likely to become intermit-
provided partial protection and elicited high antibody
tent shedders.
titers (65, 110, 449, 452). Recombinant ALVs expressing
3) Nonviremic hens regardless of immune status. These
subgroup A (60, 125, 243, 442) and ALV‐J (220, 410)
were used to provide replacements; however, up to
envelope glycoproteins have been produced that could
four generations of testing were needed to establish
have potential as vaccines to protect against horizontal
freedom, and even then infection could not be ruled
transmission. It is worth noting that congenitally infected
out (446).
chicks are immunologically tolerant and, thus, cannot be
immunized even if a suitable vaccine was available. These Application of eradication programs of ALV to com-
chickens constitute the major source of ALV transmis- mercial flocks has depended on associations between
sion and are the most likely to develop neoplasms. virus infections in hens, eggs, embryos, and chicks (375):
(1) egg albumen may contain exogenous ALV and gs
antigen, and both are usually present together; (2) a
Treatment
strong association exists between ALV or gs antigen in
No practical measures have been found for treatment of egg albumen and ALV in vaginal swabs; (3) an associa-
the various forms of the avian leukosis complex. In tion exists between ALV in vaginal swabs or egg albumen
­general, all attempts to treat virus‐induced neoplasia and ALV in chicken embryos and newly hatched chicks.
have resulted in negative or non‐reproducible results. Consequently, hens with a low probability of producing
RNA interference (RNAi)‐based methods of inhibiting infected embryos are hens negative for virus (or gs anti-
ALV replication have been demonstrated experimentally gen) by the vaginal swab test, or hens that produce eggs
(62, 247) although its value in future treatment cannot be with albumen free from virus or gs antigen. Commonly,
predicted. Recombinant chicken interferon‐alpha can virus in vaginal or cloacal swabs may be detected by
inhibit ALV replication in DF‐1 cells and could be useful ELISA, NP, or PM tests and in egg albumen by ELISA or
for antiviral approaches (95). direct COFAL tests. It is unlikely that a single test will
detect all potential shedder hens. A problem that arises
in applying the ELISA test to albumen or swabs is the
Prevention and Control Procedures
need to differentiate positive reactions due to the pres-
Eradication ence of gs antigen derived from endogenous ALV or loci
Eradication of ALV from primary breeding stocks is the from the reactions due to the presence of exogenous
most effective means for controlling ALV infection in ALV infection. Reactions due to the latter are usually
chickens. Primary breeding companies of layer‐type and markedly higher, but the setting of the boundary between
meat‐type stock have made significant progress in reduc- endogenous and exogenous virus infections is some-
ing or eradicating ALV of subgroups A, B, and J from times difficult and somewhat arbitrary. High reactions
their elite breeding lines (306). due to exogenous virus are clearer with albumen samples
Programs for eradication of ALV infection depends on than with swabs (89).
breaking the vertical transmission of virus from dam to A procedure for eradication of ALV involves: (1) selec-
progeny. Breeder hens are tested by various methods for tion of fertile eggs from hens negative in the egg albumen
the presence of ALV, and those that test positive are dis- or vaginal swab test (reviewed in 266); (2) hatching of
carded. In order to establish an ALV‐free flock, it is nec- chicks in isolation in small groups (25–50) in wire‐
essary to hatch, rear, and maintain in isolation a group of floored cages, avoidance of manual vent sexing and vac-
chickens free from congenital infection. To achieve this, cination with a common needle to prevent mechanical
embryos must be obtained from dams that are not spread of any residual infection; (3) testing of chicks for
­transmitting virus to their progeny. In earlier work on ALV by a biologic assay or PCR on blood, discarding
Chapter 15  Neoplastic Diseases 625

reactors and contact chicks; and (4) rearing ALV‐free commercial lines of chickens (84, 260). In some lines,
groups in isolation (130, 440). In practice, selection of high frequencies of a resistant allele may be found natu-
hens with a low shedding rate is a simpler requirement to rally. In others, frequencies of the resistant alleles can be
fulfil than the subsequent chick testing and isolation increased by artificial selection.
rearing needed to achieve complete eradication. In artificial selection, genotypes of unknown parents
Consequently, some commercial breeder organizations may be determined in a progeny test by mating them to
are concentrating only on reduction of infection rate by recessive tester birds of the subgroup in question (e.g.,
hen testing. Small group hatching and rearing proce- arar for A subgroup virus) (293). Depending on the seg-
dures allowed identification and removal of groups con- regation of susceptible and resistant progeny in a par-
taining chickens infected prior to hatching and prevented ticular mating, the genotype of the unknown parent may
horizontal transmission of ALV‐A in egg‐type chickens be determined. The phenotypic identification of progeny
and ALV‐J in meat‐type chickens (440). in the test may be determined by inoculation of RSV
Chicks are most susceptible to contact infection by onto the CAM, the embryo being scored as susceptible
ALVs during the period immediately after hatching. or resistant on the basis of pock count (86) or intracra-
Although congenitally infected hatchmates are likely to nial inoculation of RSV into hatched chicks, chicks being
be the main source of such infection, several procedures scored on the basis of death or survival. The former
can reduce or eliminate infection remaining from previ- method is preferable and has many advantages.
ous populations. Incubators, hatchers, brooding houses, There are concerns whether genetic selection approach
and all equipment should be thoroughly cleaned and dis- could cause problems with generation of mutant viruses
infected between each use. Chick boxes should not be that may overcome the restriction imposed by this selec-
reused, and each farm ideally should have only one age tion (78). However, this type of resistance is poorly
group of chickens. Demonstration of natural infection defined but may be controlled by a number of genes and
and transmission of MAV‐1 in egg‐type chickens stress is, consequently, more difficult to overcome by viral
the importance of testing of birds to prevent introduc- mutation.
tion of infection (447). The danger of introducing strains Recent technological advances in genome editing have
of virus not already present in the population can be already demonstrated the feasibility of this approach in
eliminated if eggs or chicks from different sources are generating cell lines resistant to infections with different
not mixed, and if chicks are reared under isolation, con- ALV subgroups (217, 218). Success in the application of
ditions that will prevent cross‐contamination of flocks. new methodologies of avian transgenesis using geneti-
cally modified primordial germ cells (174, 271) point
Selection for Genetic Resistance toward the feasibility of using these approaches for
The frequencies of the alleles that encode cellular sus- experimental generation of resistant stock as another
ceptibility and resistance to infection by exogenous tool for the control of avian retroviral diseases in
ALSVs (see Genetic Resistance) vary greatly among poultry.

­Reticuloendotheliosis
Guillermo Zavala and Venugopal Nair

Summary Intervention.  Eradication of the pathogen by testing and


eliminating infected birds is the best intervention
Agent and Disease.  Reticuloendotheliosis (RE) represents a strategy. However, vaccination has very little value,
group of syndromes associated with a common, but not except perhaps in endangered avian species.
ubiquitous retrovirus designated reticuloendotheliosis
virus (REV). The syndromes include chronic lymphoid
neoplasia, runting disease syndrome or acute reticulum cell
Introduction
neoplasia. Reticuloendotheliosis virus also poses significant
Definition and Synonyms
danger as a contaminating pathogen in avian vaccines.
Reticuloendotheliosis (RE) designates a group of syn-
Diagnosis.  Clinical diagnosis of the disease is challenging dromes associated with chronic and acute neoplasia,
because of the difficulty in differentiating from immunosuppression, runting disease, and acute death in
other  avian neoplastic diseases. Hence virological, several avian species caused by retroviruses of the reticu-
molecular, and serological diagnostic tests are needed loendotheliosis virus (REV) group. Clinical disease is
for confirmation. infrequent but infection appears to be widespread.
626 Section II  Viral Diseases

The REV group includes the laboratory strain T Recent historical, phylogenetic, and paleovirological
(REV‐T), chick syncytial virus (52), duck infectious ane- ­evidence suggest the origin of REV as a mammalian ret-
mia virus (140), and spleen necrosis virus (230). Many rovirus that was iatrogenically introduced into the avian
other nondefective strains have been isolated from avian hosts which subsequently spread through herpesviruses
species including turkeys, chickens, ducks, pheasants, and poxviruses (77, 171).
partridges, geese, prairie chickens, sparrows, and pigeons
(87, 166, 231, 271). Nondefective strains associated with
runting disease and chronic neoplasia belong to a single History
serotype, but three antigenic subtypes have been identi-
fied (46). The acute reticulum cell neoplasia is induced The the defective strain T (REV‐T), was obtained in 1957
only by the laboratory‐derived defective strain T (REV‐T) from turkey visceral lymphomas (191). REV‐T is acutely
not known to occur in nature. REV‐T carries a unique oncogenic, causing death of young chicks 6–21 days
oncogene of cellular origin (v‐rel) that is responsible for postinoculation (207). Theilen et al. confirmed the acute
its acute oncogenicity (101, 102). Stocks of REV‐T also oncogenicity of REV‐T for young chickens, turkeys, and
contain a nondefective helper REV designated as REV‐A Japanese quail; and designated the disease as reticuloen-
that replicates in chicken fibroblasts but lacks acute dotheliosis on the basis of the prominent cell type in
oncogenic properties (101). tumors (227), now termed acute reticulum cell neopla-
sia. Importantly, Theilen et  al. (227) propagated the
Economic Significance strain in cell culture, determined it to be a retrovirus
unrelated to avian leukosis virus, and named it “reticu-
Severe runting syndrome, feathering abnormalities, loendotheliosis” virus (strain T).
chronic neoplasia, or immunosuppression have occurred Purchase (182, 183) recognized the antigenic relation-
when REV‐contaminated vaccines were administered to ships between REV‐T, the nononcogenic chick syncytial
embryos or very young chickens (85, 112, 127–129, 267). spleen necrosis and duck infectious anemia viruses, all
Clinical disease from natural infection is relatively rare, within the reticuloendotheliosis virus group. Thus,
but exports of seropositive breeding stock may be pro- nomenclature for the disease and the virus originated
hibited. Vaccine and specific pathogen free companies from the atypical pathology induced by REV‐T and was
must routinely monitor their products for REV contami- extended to all viruses in the group.
nation. Significant immunosuppression may result from Additional reviews on the history of REV are available
environmental exposure, administration of contami- for consultation (147, 166, 171, 177, 183, 249).
nated vaccines, or congenital transmission (250, 267).

Public Health Significance Etiology


The extended host range of REVs, which includes certain
Classification
mammalian cells (2, 171, 243) and other characteristics
of REV suggesting an evolutionary linkage with mamma- Reticuloendotheliosis viruses are retroviruses that are anti-
lian retroviruses (15, 126, 171, 189), raised the possibility genically, morphologically, and structurally distinct from
of human infection (113). Reticuloendotheliosis virus the leukosis/sarcoma (L/S) group of avian retroviruses
antibodies have been detected in human sera (49, 114, (ALSV) (183). Lack of nucleic acid sequence homology
115), but such findings have been considered insufficient between REV and members of the ALSV group has been
to warrant concern (68, 70, 71, 94, 200). long recognized (118). The International Committee on
Taxonomy of Viruses (ICTV 9th Report, 2011) has classi-
fied REVs within the family Retroviridae, subfamily
Scientific Significance
Orthoretrovirinae, genus Gammaretrovirus, with no
Reticuloendotheliosis virus has received considerable endogenous counterpart. Reticuloendotheliosis virus is
attention as an oncogenic and immunosuppressive virus phylogenetically related to mammalian C‐type retroviruses
with a wide host range (171). Reticuloendotheliosis virus based on virion morphology, nucleic acid and major poly-
can infect or transform various cell types (2, 15) and has peptide amino acid sequences (171), as well as immuno-
been used in comparative retrovirology models. Some logic determinants and receptor interference patterns (122).
REV subgenomic sequences display high similarity to
those of some mammalian retroviruses (17, 171).
Morphology
Reticuloendotheliosis virus can integrate into the
genome of cells and of large DNA viruses, including Viral particles are typical of retroviruses, with about
Marek’s disease (MD) and fowl pox viruses (100, 110). 100 nm in diameter (270); display surface projections
Chapter 15  Neoplastic Diseases 627

about 6 nm long and 10 nm in diameter (116). Virions Oncogene


have a density of 1.16–1.18 g/mL in sucrose density The v‐rel oncogene is transcribed in REV‐T‐transformed
­gradients (23), and can be differentiated from avian L/S lymphoid cells and produces a phosphoprotein product
viruses by morphology in thin sections (153, 272). identified as pp59v‐rel. The v‐rel protein is a member of the
The  morphology of the viral particles is shown in rel/dorsal family of proteins, related to nuclear factor kappa
Figure 15.46. B, which function as DNA‐binding transcription factors (26,
194). It differs from the c‐rel protein both in structure and
transforming ability and, unlike most other oncogene prod-
Chemical Composition
ucts, can be detected in both the cytoplasm and nucleus of
Nucleic Acid transformed cells (29). The v‐rel protein is usually com-
The genomic single‐stranded, positive sense RNA con- plexed with cellular proteins (124, 136, 216) and is responsi-
sists of a 60–70 S complex containing two 30–40 S RNA ble for the acute oncogenicity of REV‐T (29). REV‐T‐induced
subunits, each having a size of about 3.9 × 106 d (25, 142). transformation is associated with several changes in gene
The nondefective REV has a genome of about 9.0 kb expression, including induction of miRNAs (264). However,
(17), while the replication‐defective REV‐T genome is REV isolates other than REV‐T have induced neoplastic dis-
only about 5.7 kb due principally to a large deletion in ease within very short latent periods (18, 72, 73, 184, 186).
the gag‐pol region and a smaller deletion in the env
region (50). Moreover, the REV‐T genome contains a Proteins
substitution of 0.8–1.5 kb in the env region that repre- Reticuloendotheliosis virus genes encode various struc-
sents the transforming gene v‐rel (44, 51, 261), which is tural proteins, a protease, a polymerase and an integrase
not present in nondefective REVs or other avian or (17). The protein encoded by the v‐rel gene is only pre-
mammalian retroviruses. Related sequences (c‐rel sent in the defective REV‐T. The RNA‐dependent DNA
proto‐oncogene) are present in the DNA of normal polymerase (reverse transcriptase) differs structurally
avian cells, including turkey cells from where the onco- and immunologically from comparable enzymes of
gene was most likely transduced (44, 45, 247, 248). No l­eukosis/sarcoma viruses (22, 153). The preference of the
endogenous REV sequences in host DNA have been rec- REV polymerase for Mn2+ ions is a differentiating factor
ognized. The long terminal repeats (LTRs), 569 base from enzymes of other avian retroviruses (153, 203, 263).
pairs (bp) in length (17) are efficient promoters in a vari- The envelope protein is composed of two peptides, the
ety of cell types (14, 17, 138). Several complete genome gp90 surface unit (SU) and the gp20 transmembrane unit
sequences of various field and vaccine‐contaminant iso- (TU) (233, 234). These glycoproteins are located on the
lates from China and the United States have been surface of the virions (156). The C‐terminal epitope
resolved (14, 17, 138). of  gp90 locates on the surface of infected cells (234).

(A) (B)

Figure 15.46  Electron micrographs of thin sections of chicken embryo fibroblasts infected with reticuloendotheliosis virus (REV). (A)
Typical virus particles in the extracellular spaces ×40,000. (B) REV particles budding from the plasma membrane of infected cells (arrow).
×60,000. (Nazerian).
628 Section II  Viral Diseases

The  gp90 protein is considered the immunodominant Host Range


protein of the virus (64). The receptor binding regions Cell cultures from many avian species and certain mam-
have been mapped and display structural differences malian cells are susceptible to infection and at least limited
relative to other retroviruses (144). viral replication. Nondefective REV has been grown in D17
There are five gag gene‐encoded structural proteins, dog sarcoma cells (15, 242, 243), Cf2th dog thymus cells (2,
p12, pp18, pp20, p30, and p10 (232). Antiserum to the 209), normal rat kidney cells (121), mink lung cells (2), and
30 kDa (p30) protein cross‐reacted with p30 of several other mammalian cells. Rat and mouse cells were only
other REVs, thus establishing this protein as a group‐ semi‐permissive for replication of REV (75, 76). Chimeric
specific antigen (143) that also plays a role in viral parti- vector particles containing the REV‐A matrix protein
cle assembly and encapsidation (244). infected mammalian cells more efficiently than those con-
taining the matrix protein of spleen necrosis virus (43). A
wide range of avian species support REV replication in vivo
Replication
but there is little evidence for in vivo replication of REV in
Non‐Defective Strains non‐avian species. Presence of REV antibodies in humans
In vitro virus replication is similar to that of other retro- has been reported (114) but the significance is unknown at
viruses (70). The virus envelope glycoprotein binds to an best (200). Although REV‐A‐based vectors could infect
unidentified cell surface receptor, resulting in interfer- human cells (125), REVs did not infect human cells due to
ence with superinfection (86). Viral entry, RNA reverse inability to bind to specific receptor (94).
transcription and proviral DNA integration into the cel-
lular chromosome proceed through mechanisms typical Pseudotypes
of simple retroviruses. Viral RNA transcription and The envelope of nondefective REV forms pseudotypes
translation are initiated through promoter and enhancer with Rous sarcoma virus (198, 235) and with vesicular
sequences in the LTR. Two polyproteins are encoded, stomatitis virus (117). Pseudotypes can be neutralized by
gag‐pro‐pol and env; the gag precursor protein is myri- REV antiserum, and thus this assay was once used for
stylated. The encapsidation sequence is located in the antibody detection (56).
gag gene. The final stage is budding of viral particles
from the cell membrane. Virus particle production is Insertional Mutagenesis
first noted at 24 hours (116), and peaks 2–4 (101) days Replication of REV requires integration of proviral DNA
after infection in chicken cells (30, 90, 223, 224). into the host cell genome. REV proviral DNA can also
integrate into the genomes of high molecular weight
Defective Strain DNA viruses including MD virus (111) and fowl poxvirus
The defective REV‐T requires a nondefective RE helper (92, 100). Insertions occur both in vitro and in vivo (60)
virus for replication (101). Oncogenicity of this strain is and may result from coinfections of REV and a recipient
maintained during passage in vivo (191) or during cul- DNA virus. Most insertions consist of a solitary, partially‐
ture of infected hematopoietic cells (101), but is rapidly deleted LTR (60, 154). However, full‐length, infectious
lost during passage in fibroblast (227, 257) and dog REV genomes have been detected in turkey herpesvirus
­thymus cell cultures (2), possibly due to the loss of the (110). A nearly full‐length, infectious REV provirus has
replication‐defective, acutely oncogenic REV after serial been detected (92, 100, 123, 210) in certain strains of fowl
passages (31). poxvirus, even in fowl poxvirus stocks lyophilized for
over 50 years (123). Such insertions could alter the
Cytopathology ­biological properties of the recipient organism and also
Replication of REV in avian fibroblasts may induce sub- represent a distinct mechanism of infection.
tle cytopathic changes (227), such as syncytia (52) but
degenerative changes are more commonly seen (223,
Strain Classification
225). Accumulation of unintegrated viral linear DNA in
infected cells possibly causes cell death. Cells that are The antigenic properties of different REV isolates are
able to prevent early superinfection have few copies of remarkably uniform (33, 182) and, except for defective
unintegrated viral DNA and survive (225). REV‐T, REV isolates have similar genetic, structural, and
The acute cell death phase (Figures 15.47A and 15.47B) chemical properties (22, 119). Although REVs belong to
lasts 2–10 days postinfection and is followed by a chronic a single serotype (46), three subtypes were identified on
infection state without cytopathology but with contin- the basis of neutralization tests and differential reactivity
ued virus production (Figure  15.47C) (223, 224). This with monoclonal antibodies (46, 57), although subtypes
cytopathology is the basis of plaque assays (35, 47, 48, 1 and 2 could not be differentiated by receptor interfer-
155, 223), but the methods have not been widely used ence (86), confirming the absence of major differences
due to inconsistency in cytopathology. between subtypes. Reticuloendotheliosis virus isolates
Chapter 15  Neoplastic Diseases 629

differ also in certain biologic properties, including path- Cell Lines


ogenicity (183) and replication in vivo. Hematopoietic cells transformed in vivo or in vitro by
REV‐T have been developed into continuous cell lines;
the cell types and surface markers vary based on the
Laboratory Host Systems
strain of helper virus and on whether transformation
Cell Cultures occurred in vivo or in vitro (29, 105). A line of trans-
Fibroblasts from several avian species and certain cell formed chicken embryo fibroblasts has also been devel-
lines, such as QT35 quail sarcoma cells (48, 54) and D17 oped (89). Cell lines have also been derived from chronic
dog osteosarcoma cells (15, 243), are susceptible to infec- lymphomas induced by nondefective REV strains (168,
tion with nondefective REVs. In infected cultures, anti- 184). Cell lines induced by in vitro transformation of
gens (Figures 15.47B and 15.47C), virus particles, proviral spleen cells with defective REV are useful expression
DNA, cytopathology, and reverse transcriptase may be ­systems for transfected foreign genes (181, 202) or as
detected and serve as criteria for virus assays. A quanti- substrates for the propagation of other viruses (187).
tative fluorescent focus assay has been developed for Some of these transformed cell lines produce growth
infected cultured cells overlaid with agar (182). Duck ­factors or cytokines (69, 93, 95).
embryo fibroblasts are preferred for demonstration of
cytopathic effects (13). However, virus cultures in
chicken embryo fibroblasts or DF‐1 cells combined with
Pathobiology and Epidemiology
molecular detection of REV or identification of viral pro-
teins by direct or indirect immunofluorescence appear
Incidence and Distribution
to be most practical and efficient (17, 18).
Reticuloendotheliosis virus infection is common but not
Embryos and Birds ubiquitous in turkeys, ducks, and chickens. The preva-
Laboratory host systems for REV that are now seldom lence of seropositive flocks and of seropositive birds
used include chicken embryos (208) and a variety of within an infected flock both increase with the age of the
avian species including young chickens, Japanese quail, flock. A high prevalence of virus or antibodies has been
ducks, geese, turkeys, pheasants, and guinea fowl (18, detected in the United States, Japan, Korea, and Egypt (4,
186, 227). 7, 13, 206, 237, 262). Seropositive commercial chicken

(A) (B) (C)

Figure 15.47  Acute (cytopathic) and chronic (noncytopathic) infection of chicken embryo fibroblasts inoculated with nondefective
reticuloendotheliosis virus (REV) strain. (A) Mild cytopathic changes 13 days after infection. Unstained, ×55. (B) Cytopathic changes and
viral antigens 13 days after infection demonstrated by indirect immunofluorescent staining. (C) Chronically infected cultures 48 days after
infection showing relatively normal‐appearing cells, most of which contain cytoplasmic viral antigens. ×360.
630 Section II  Viral Diseases

flocks are still common in the United States, with sporadic infestans and Ornithodoros moubata after feeding on
to negligible clinical disease. Reticuloendotheliosis virus infected chickens (62, 228, 229) and for up to five hours
infection is a significant clinical problem in wild endan- post‐feeding on contaminated blood in some species of
gered avian species (17, 74, 87, 268). Reticuloendotheliosis mosquitoes (62). Reticuloendotheliosis virus has also
virus is frequently detected alone or in combination with been isolated from mosquitoes (Culex annulirostris)
fowl poxvirus or lymphoproliferative disease virus in wild (164) in contact with viremic chickens, demonstrating
turkeys of the United States (1, 3). the possibility of mechanical transmission by insects,
Runting disease syndrome and chronic neoplasia which may explain seasonal seroconversion (63, 164) and
have occurred following vaccination with REV‐contam- a higher prevalence of infection in Southern states (254,
inated vaccines (13, 85, 112, 120, 185, 265, 266). 256). Fowl poxvirus, which is also transmitted by mos-
Immunosuppressive disease has been identified after quitoes, may function as a vector containing infectious
natural infection in Korea (206). Field cases of RE‐ clones of REV (92, 100, 211).
related lymphomas in turkeys have been described in
the United States (55, 175, 215, 252, 258, 259), England Vertical Transmission
(147), and Israel (107). Losses from mortality and con- Chickens, turkeys, ducks, and quail with persistent
demnation at slaughter in affected flocks could be as viremia may transmit infectious REV to progeny,
high as 16–20% (146, 175). Lymphomas associated with although usually at low frequency or not at all (10, 12, 18,
natural REV infection are sometimes reported in wild 148, 236, 260). However, some studies have reported
turkeys (99, 132) and less often in chickens (61, 107, 150, experimental vertical transmission to over 50% of chicks
173). Chronic REV‐induced neoplasia has also been from infected dams (163). Albumen samples from toler-
occasionally observed in ducks (97, 176), quail (39, 201), ant hens frequently contained RE viral gs antigen,
pheasants (73), geese (72), peafowl (152), and prairie although infectious virus was rarely isolated (260).
chickens (74, 268). Vertical transmission may occur at higher rates in ducks,
since virus was isolated from 87% of embryos derived
from tolerant females (158). Individual antibody‐­
Natural and Experimental Hosts positive, virus‐positive turkey hens may still transmit
virus to progeny at a high rate (258).
Natural hosts for REV infection include turkeys, chick-
Semen from tolerant turkeys contains infectious virus
ens, ducks, geese, pheasants, Japanese quail, peafowl,
(149, 257). Reticuloendotheliosis virus‐free turkey hens
and prairie chickens. Experimental hosts include most of
inseminated with contaminated semen have produced
the above species, as well as guinea fowl, chickens,
infected progeny (148). There is evidence of limited
­turkeys, and Japanese quail (18).
vertical transmission after mating infected males with
noninfected female chickens (195). Genetic transmis-
sion is unlikely based on lack of clonal insertions of pro-
Transmission, Vectors, Carriers
viral DNA in infected hens or their progeny (258).
Horizontal Transmission Iatrogenic infection is possible by using accidentally
Experimentally, REV can be transmitted by close contact contaminated needles or vaccines for embryos or very
with infected chickens, turkeys, and ducks (130, 158, 174). young chickens (13).
Horizontal transmission may be influenced by the
host  species (182, 254) and the virus strain (254, 266). Contaminated Biological Materials
Reticuloendotheliosis virus transmission was not detected Accidental use of REV‐contaminated fowl pox (85, 138)
when chickens were separated by wire mesh (12). or MD (112, 120, 134, 135, 138, 245, 266) vaccines
Many flocks become infected at older ages (254) pre- has  been documented. Reticuloendotheliosis virus has
sumably via contaminated environment, insects, and also been detected in contaminated vaccines against
other biological reservoirs. Reticuloendotheliosis virus Newcastle disease and infectious bronchitis (245).
has been detected in feces and cloacal swabs (10, 179, Certain stocks of avian myeloblastosis virus, for many
260, 266), body fluids (12), and litter (241). Horizontal years distributed as a source of reverse transcriptase for
transmission was limited amongst experimentally biochemical purposes, contained a low level of REV
infected Japanese quail housed on litter (18). Flocks (256). Quality control procedures to exclude REV from
infected experimentally (Zavala, unpublished) or natu- licensed poultry biologics have not always been uni-
rally at later ages seroconvert, making virus detection formly effective for detection of REV contamination in
difficult (255). Furthermore, REVs are quickly degraded vaccines such as fowl pox (78, 84). Subgenomic REV
outside the host at ambient temperatures (37). sequences continue to be found in some (67, 92, 100,
Insect transmission represents another form of hori- 210), but not all (154), commercially produced fowl pox
zontal spread. Virus could be recovered from Triatoma virus vaccines. Reticuloendotheliosis virus has also been
Chapter 15  Neoplastic Diseases 631

detected in stocks of Plasmodium lophurae (140, 230), c­ linical signs due to the rapid onset of the disease, and
further illustrating the diversity of possible transmission mortality rates often reach 100% (208, 227).
mechanisms for REV.
Pathology
Incubation Period
Runting Disease Syndrome
The runting disease syndrome represents a non‐neo- In chickens, the principal changes include runting (165,
plastic disease process with an outcome depending on 257), thymic and bursal atrophy (165), enlarged peripheral
virus strain and other factors. Atrophic changes in the nerves (257), abnormal feathering (112, 127–129), proven-
bursa and thymus can be seen as early as three days triculitis (112), enteritis (146), anemia (120, 140), and
postinfection (259). Persistent weight depression in hepatic and splenic necrosis (183, 230), often accompa-
infected chicks can be detected as early as six days of age nied by depression of cellular and humoral immune
(159). By the second week postinoculation, chickens responses (17, 32, 40–42, 107, 120, 255). The acute hem-
developed microscopic nerve lesions (257) and had orrhagic or chronic ulcerative proventriculitis observed in
depressed immune responses (255). Japanese quail field cases (112) could not be reproduced (11) with a simi-
infected experimentally displayed severe weight depres- lar isolate. The proliferative lesions in enlarged peripheral
sion as early as 14 days of age and lymphomas were nerves often occur in the absence of other neoplasms and
detected as early as 35 days of age (16). it is not known whether the changes are neoplastic or
Chronic neoplastic responses occur after moderate or inflammatory (257). The infiltrating cells, which include
long incubation periods. Chickens developed bursal‐ lymphocytes and plasma cells, are shown in Figure 15.48.
derived B‐cell lymphomas 17–43 weeks after inoculation Ducks inoculated with the spleen necrosis or duck
(260). Reticuloendotheliosis virus‐associated lymphomas infectious anemia strains of REV may display some fea-
in turkeys occurred between 15 and 20 weeks of age in tures of the runting disease syndrome (140, 230).
some trials (146, 175) and as early as 9 weeks post‐infection Hematocrit values in ducks inoculated with spleen
in SPF turkeys (Zavala, unpublished). In transmission stud- necrosis strain can be as low as 20%, compared to 35%
ies, lymphomas were induced after 8–11 weeks (174) or for control ducks (230). Enlarged nerves (174, 175) or
11–12 weeks (146). Chronic lymphomas occur between 20 enteritis (146) have been observed in turkeys with RE‐
and 30 weeks in the domestic goose (72), and at 4–24 weeks related chronic lymphomas.
in ducks (97, 176, 178). Experimental inoculation of newly Genetic differences in susceptibility have not yet been
hatched ducks induced lymphomas and other neoplasms described; chicks from lines of different susceptibility to
between 8 and 30 weeks (133, 158). MD were equally susceptible to the development of
For acute reticulum cell neoplasia, the incubation nerve lesions following inoculation with REV (257).
period can be as short as 3 days, but death occurs more Most nondefective REV strains, when inoculated at
commonly 6–21 days after inoculation (207). Because of hatch, induce high frequencies of gross lesions (182, 255)
the short latent period and high mortality induced by
REV‐T, this virus has been regarded as the most virulent
of all retroviruses (29).

Clinical Signs
Chickens and quail with runting disease syndrome may be
notably stunted and pale (16, 165). Weights of infected
chickens and quail may be 20–50% lower than controls by
3–5 weeks after infection (16, 255, 257). Weight depression
has also been seen in infected ducks (182). Some chickens
may display abnormal feather development, termed
Nakanuke, that is, wing feathers with focal adhesion of the
barbs to the shaft (127, 128). Lameness or paralysis is rare
even in birds with gross nerve lesions. Mortality might be
rare in chickens (255) but affected birds in commercial
flocks may be culled prior to death; a culling loss of over 50%
between 5 and 8 weeks was described in one flock iatrogeni-
Figure 15.48  Microscopic lesions in a peripheral nerve of a
cally infected with a ­contaminated MD vaccine (221). chicken inoculated with nondefective reticuloendotheliosis virus
Birds developing chronic lymphomas or acute reticu- (REV) strain. Infiltrating cells consist of mature and immature
lum cell neoplasia after REV‐T infection show few lymphocytes and plasma cells.
632 Section II  Viral Diseases

but others, such as chick syncytial strain, may induce not always be present in field cases. Grimes et  al. (96)
few, if any, lesions (255). observed what may be lymphomas in two chickens at 22
and 24 week after inoculation with a field strain of REV,
Chicken Bursal Lymphoma but no bursal involvement was reported. However, typi-
Chickens inoculated with the nondefective chick syncyt- cal bursal lymphomas were observed in two chicken
ial or T strains developed B‐cell lymphomas, involving flocks following administration of a REV‐contaminated
principally the liver and bursa of Fabricius (251, 260). fowl pox vaccine (85).
The gross lesions were nodular or diffuse lymphoid
lesions in the liver, other visceral organs and the bursa of Chicken Non‐Bursal Lymphoma
Fabricius, all indistinguishable from lymphoid leukosis Chronic non‐bursal lymphomas have been described in
(Figure  15.49). A few birds may develop sarcomas or chickens following experimental infection with the
adenocarcinomas. The frequency of lymphomas was
­ spleen necrosis or chick syncytial strains of REV (259).
influenced by virus strain and whether a tolerant infec- Grossly, these lymphomas are focal or diffuse lymphoid
tion had been induced (260). Interestingly, coinfection of infiltrations, with enlargements of the thymus, liver, and
chickens with serotype 2 MDV enhanced the incidence spleen or focal lesions of the myocardium (Figure 15.50).
of REV bursal lymphomas (6) as had also been reported The bursa of Fabricius is not involved. Nerve enlarge-
for lymphoid leukosis (8). ments may be seen. Histologically, the tumors appear to
The tumor cells, which are uniform populations of be a uniform, immature lymphoreticular cell that lacked
lymphoblasts, were identified as B cells by IgM and B‐cell markers and did not express MATSA, a cellular
other  B‐cell specific markers (168, 260). The bursal antigen associated with MD tumors (259) and also
dependency of this tumor was confirmed in chemically expressed on activated T lymphocytes (145). The princi-
or surgically bursectomized chickens, which were refrac- pal tumor cell type is a CD8+ T cell but Ia antigens are
tory to tumor development (82). Bursal lymphomas may not expressed (53).

Turkey Lymphoma
Chronic lymphomas in turkeys and other avian species
consist of gross lymphoid infiltrations in the liver, intes-
tine, spleen, and other viscera. Lymphomatous lesions in
the bursa have been reported (146, 174), but this lesion
was not common. Histologically, the lesions were com-
posed of uniform populations of lymphoreticular cells

Figure 15.50  Nonbursal lymphoma in a chicken 48 days


postinoculation with the nondefective spleen necrosis strain of
reticuloendotheliosis virus (REV). Note enlargement of spleen,
Figure 15.49  Bursal lymphoma in a chicken. Gross lymphomas in nodular lymphomas on heart, and bursal atrophy of infected
the liver and bursa of a chicken 25 weeks after inoculation with chicken (top row). Organs from age‐matched controls in the
the nondefective chick syncytial strain of REV. bottom row (262).
Chapter 15  Neoplastic Diseases 633

(146, 174). Crespo et al. (55) described T‐cell lymphomas


associated with a natural outbreak of reticuloendothelio-
sis in turkeys.

Lymphomas of Other Species


Various other species develop chronic gross lympho-
matous lesions that are similar to those described for
chickens and turkeys. Lesions reported in ducks
include enlarged livers and spleens with diffuse or
focal involvement, intestinal lesions, and infiltrations
in skeletal muscle, pancreas, kidneys, heart, and other
tissues (97, 133, 158). Generalized leukemia in addi-
tion to visceral lymphomas in ducks has been described
(178). Similar tumors were described in geese, with
occasional lymphoproliferative lesions in the bursa of
Fabricius (72). Pheasants and prairie chickens dis-
played proliferative cutaneous lesions on the head and Figure 15.51  Microscopic lesions of acute reticulum cell neoplasia
mouth in addition to nodular visceral lymphomas (73, (reticuloendotheliosis) in the liver of a chicken inoculated with
74, 268). Outbreaks in quail were characterized by liver replication‐defective, acutely transforming strain T REV. The liver is
infiltrated with large primitive reticular cells (arrow).
and spleen enlargements (16, 18, 39, 201, 268) or intes-
tinal lesions (268). Histologically, tumors from all these
species generally resembled those described for chick- Persistent infections occur rarely following inoculation
ens and turkeys. at hatch (10, 148, 260) depending on the strain of
chicken (83); and are unlikely to occur if exposure
Acute Reticulum Cell Neoplasia occurs at later ages. Some birds with persistent infec-
The pathology of acute reticulum cell neoplasia has been tions develop antibody responses. Tolerant infection is
well described (191, 227). Grossly, affected birds develop associated with higher rates of vertical transmission
infiltrative focal or diffuse lesions in the livers, spleens, and tumor development, and birds are typically stunted
pancreas, gonads, heart, and kidney. The blood shows a and immunodepressed. Birds exposed after hatch most
decrease in heterophils and an increase in lymphocytes commonly develop transient viremia followed by anti-
(222), leading to leukemia a few hours before death. body production (10, 255). Persistence of noninfectious
Histologically there is infiltration and proliferation of RE viral antigens in the blood for several weeks follow-
cells described either as large mononuclear cells of the ing the disappearance of infectious virus has been
reticuloendothelial system (227) or primitive mesenchy- reported (10). Transient infection rarely results in
mal cells (191, 208). Some lesions are composed almost ­vertical transmission, immunosuppression, or tumor
solely of such cells, whereas others include also smaller development. Infection of older birds rarely results in
lymphoid elements, probably indicating a host immuno- clinical disease (179, 182, 254, 266) except, perhaps, in
logic response. Areas of necrosis are also frequent. turkeys in which lymphomas have been observed
A typical liver lesion is shown in Figure 15.51. ­following contact exposure (148, 149, 174).
Various other factors influence susceptibility to infec-
Multiple Syndromes tion or disease. No genetic cellular resistance has been
Lesions of different types can be observed in the recognized. However, some differences in the pathologic
same  flock, experiment, or even in individual birds. response of genetic lines or families has been recognized
Nondefective REV strains may first induce runting in chickens (83, 205, 259) and quail (226). Nevertheless,
­disease syndrome and lymphomas may occur later in the differences were not apparent when two different chicken
survivors, sometimes accompanied by peripheral nerve lines were challenged with serial dilutions of REV‐T
enlargement. (207). Maternal antibodies appear to limit susceptibility
to infection (213).
Pathogenesis
Runting Disease Syndrome
Virus Infection The pathogenesis of runting disease syndrome has not
Tolerant infection with persistent viremia and absence been elucidated. Stunted chickens did not consume less
of antibodies is readily induced in turkeys (148, 258) feed, but had marked reduction of phosphoenolpyruvate
and chickens by embryo inoculation (107, 260); and carboxykinase, a key gluconeogenic enzyme in the liver
by  vertical transmission from infected dams (12). (93). The adherence of feather barbules to the shaft
634 Section II  Viral Diseases

(Nakanuke) is apparently due to REV‐induced necrosis REV‐T. Transformation does not require the presence of
of feather‐forming cells (219). The microscopic lesions a helper virus (131). Lymphoid cells transformed by
of chicks with runting syndrome resemble a graft versus REV‐T in vitro, but which produce no infectious virus,
host reaction (165), but a specific autoimmune compo- will produce typical RE when transplanted into synge-
nent has not been identified. neic recipients (131, 193).

Chronic Lymphomas
Immunity
The integration site for the REV DNA provirus in the
cellular genome in bursal lymphomas is located adjacent Humoral Responses
to c‐myc, a cellular oncogene important in the induction Birds with nontolerant infections develop robust anti-
of lymphoid leukosis by avian leukosis virus (ALV) (172). body responses, detected as early as 16–21 days after
The molecular mechanism by which c‐myc is activated inoculation in chickens (32, 157), but 6–10 weeks may be
by insertion of REV proviral DNA has been described required in contact‐exposed birds (107, 130, 148).
(91, 190, 218). The proviral insert often contains major Antibody titers may decline with age (10, 32, 260), but
deletions that prevent the expression of infectious virus neutralizing antibodies have been detected in experi-
(217). Based on pathology, proviral insertional activation mentally‐infected turkeys through 40 weeks (148). Most
of c‐myc, and enhancement by serotype 2 MDVs, bursal birds that develop tolerant infections do not develop
lymphomas induced by REV, and ALV appear indistin- humoral immune responses, although a few tolerant
guishable. However, some subtle differences have been chickens ultimately develop antibodies (159). The pres-
noted. For example, chickens of lines resistant and ence of antibodies may influence tumor susceptibility as
­susceptible to lymphoid leukosis were uniformly less chemically bursectomized quail were more susceptible
susceptible to lymphoma induction by REV than by ALV to tumors than controls (186).
(83), and the REV lymphomas frequently require longer
latent periods than those induced by ALV. Cellular Responses
For nonbursal lymphomas, the molecular mechanism Major histocompatibility complex‐restricted cytotoxic-
of oncogenesis also involves insertional activation of ity against lymphoblastoid cell lines transformed with
c‐myc, but the strong tendency for the provirus to be ori- defective REV has been described in chickens within
ented in the same direction as c‐myc in bursal lympho- seven days after inoculation with defective or nondefec-
mas was not observed in nonbursal lymphomas (109). tive RE viral strains (141, 246). This response appears to
It is not known whether a common mechanism exists be mediated by activated (MHC class II+) CD8+ T cells
for oncogenesis in chronic lymphomas of chickens and (126). However, natural killer (NK) cells were not acti-
turkeys. In ducks, the frequency of experimentally‐ vated (199). The induction of cytotoxic T cells by REV
induced REV lymphomas was not affected by embryonal has been used as a general indicator of immune response
bursectomy (133), indicating that these tumors may not in the study of other avian viruses (187).
necessarily be of B‐cell origin.
Immunodepression
Acute Reticulum Cell Neoplasia Humoral and cellular immune responses are frequently
The target cell transformed in vivo by replication‐­ depressed in chickens infected with nondefective REV
defective REV‐T (with REV‐A helper virus) expresses T strains. Depressed antibody responses to MDC and tur-
lymphoid and myeloid markers (19). These cells also key herpesvirus (HVT) (32, 120), Newcastle disease
express surface MHC class I and II antigens, as well as virus (107, 265), sheep erythrocytes, and Brucella abor-
interleukin‐2 receptor (103), and are immunoglobulin M tus (255) are documented. The magnitude of antibody
(IgM) negative but vary in expression of CT3 (19). depression is influenced by the dose and strain of virus,
Similar tumors were induced in chemically bursecto- and primary responses are more severely affected than
mized chickens (19). Inoculation directly into the thy- secondary (255). Depressed responses against Pasteurella
mus induced thymomas composed of T and B cells (29). multocida in turkeys infected with nondefective REV
On the other hand, REV‐T, when associated with chick isolated from Attwater’s prairie chickens have been
syncytial virus helper virus instead of REV‐A, induces ­documented (17).
IgM‐positive B‐cell lymphomas with rearrangements of Different strains of nondefective REV varied in their
the heavy‐ and light‐chain immunoglobulin loci (20, 21). ability to induce bursal atrophy and suppression of B cell
Thus, cell tropism appears to be determined, in part, by populations available for transformation by v‐rel (20).
differential effects of the helper viruses on lymphoid Studies on chimeric viruses derived from REV‐A and
populations. chick syncytial virus showed that regions in both gag and
Neoplastic transformation in acute reticulum cell neo- env genes were associated with the strong immunode-
plasia is mediated by the oncogene v‐rel, present in pressive ability of REV‐A (88).
Chapter 15  Neoplastic Diseases 635

Spleen cells from chickens infected with REV‐T dis- DNA in tumor cells has diagnostic value. Diagnostic
played suppressed responses to the mitogen phytohe- techniques have been reviewed by Zavala et al. (269).
magglutinin (40, 204). This effect is associated with the
nondefective helper virus in strain T stocks (34, 42) and
Isolation and Identification
is mediated through a population of suppressor cells (41,
192), which could be demonstrated only through the Reticuloendotheliosis virus viremia is typically low titered
third week after infection (193). Other cellular immune and transient, except in tolerant birds. Birds with lesions
responses inhibited by REV infection include mixed are normally a good source of virus, which may be isolated
­lymphocyte reaction and allograft rejection (240). by inoculation of susceptible tissue cultures with tissue
Depression of humoral responses and a transient suspensions, blood, plasma, splenocytes, white blood
mitogen responsiveness have been identified following cells, or other inocula. Cellular inocula are preferred over
infection with the chick syncytial strain, but persisted cell‐free inocula, because of higher titers. Cytopathic
through 10–19 weeks in tolerant chickens infected with effects in cell cultures may not be evident, thus, cultures
nondefective REV (255, 260). Infected chickens were should be maintained through at least two blind 4–7‐day
more susceptible to the development of a MD tumor passages of infected cells. Replication of REV may be con-
transplant (34); to reactions from infectious laryngotra- firmed by demonstration of viral antigen in cell cultures
cheitis vaccine (160, 212); to natural fowl pox virus using fluorescent polyclonal or monoclonal antibodies
infection (162); to infectious bronchitis virus (212); to (17, 18, 57), immunoperoxidase (35), complement fixation
mortality induced by Eimeria tenella (161); and to (214), enzyme immunoassay (58, 108, 170), or molecular
Salmonella typhimurium infection (160). No increase in detection of RNA or DNA (5, 17, 38, 74, 78, 87, 92, 196,
susceptibility to MDV was noted (33), but there was 268). In comparative studies, enzyme immunoassays were
interference by REV infection with immunity induced more sensitive than complement fixation tests (58) and
by turkey herpesvirus against MD in chickens (255). indirect immunofluorescence was more sensitive than
Humoral immunodepression was also seen in ducks indirect immunoperoxidase or immunoelectron micros-
infected with a field isolate of REV (133). In the field, copy (169). A convenient and sensitive indirect immuno-
immunodepression is probably the most important con- fluorescent assay conducted in 96‐well plates (46) has
sequence of embryo‐ or vaccine‐derived REV infections been used for virus isolation from field samples (258).
but is less likely to result from contact infection (254) Virus isolated by any of these procedures may be used
and has not commonly been associated with ­seropositive for reproduction of the disease or for further tests. Yolk
flocks. Reticuloendotheliosis virus‐induced immuno- inoculations in 5–7‐day‐old chicken or quail embryos
suppression has been reviewed (267). are useful for disease reproduction and virus replica-
tion (16, 18). Virus isolates may be assigned to antigenic
Tumor Immunity subtypes by the differential reactivity to fluorescent
Regression of strain T‐induced wing‐web tumors was mAbs (46).
partially abrogated by bursectomy, thymectomy, and Detection of proviral DNA by PCR assays has been
bursectomy–thymectomy (137). Serum from hyperim- shown to be a sensitive and specific method for detec-
munized (104) chickens was protective against tumor tion of REV in chicken embryo fibroblasts or DF‐1 cells,
development even after absorption to remove antiviral paraffin‐embedded tissues, as well as in blood and
antibodies (104), suggesting the existence of tumor‐­ tumors of infected birds (5, 17, 38, 74, 78, 87, 92, 196,
specific transplantation antigens on RE tumor cells. 268). Polymerase chain reaction is useful for tumor
Chickens immunized with purified or inactivated prepa- diagnosis (59, 61, 63, 65) and for evaluating vaccines for
rations of nondefective strain T helper virus were resist- possible REV contamination (78, 84, 85, 100, 139, 220).
ant to challenge with acutely transforming REV‐T Veterinary Services Memorandum 800.88 by USDA/
preparations (24). However, immunization with empty APHIS describes PCR, in vivo virus amplification, and
virions (151) did not provide protection. serology as suitable direct or indirect tests for detection
of REV in master seed viruses for the production of
commercial poultry vaccines. Assays amplifying REV
Diagnosis envelope and REV 3’ LTR sequences provided a more
accurate assessment of REV provirus than PCR assays
A diagnosis of RE requires not only the presence of typi- that amplify the REV 5’ LTR region (92). Although PCR
cal gross and microscopic lesions, but also the demon- assays are sensitive and specific they may not be as well
stration of REV, REV antibodies and the exclusion of suited as enzyme immunoassays for large‐scale testing.
other oncogenic agents. Because REV, unlike avian leu- A loop‐mediated isothermal amplification (LAMP)
kosis and MD viruses, is not yet as ubiquitous, the dem- method for rapid detection of REV with high sensitivity
onstration of infectious virus, viral antigens, and proviral and specificity has also been reported (66).
636 Section II  Viral Diseases

Serology load by quantitative PCR analysis (188) or real‐time


PCR. Non‐quantitative PCR assays are probably of little
Serological confirmation of REV exposure involves the
value for diagnosis of MD because of the potential to
detection of antibodies in sera from experimentally inoc-
detect MDV DNA in the absence of lymphomas.
ulated chickens or from clinically affected chickens.
Polymerase chain reaction has been shown to detect
The  most sensitive test for detection of antibodies to
REV‐LTR sequences from lymphomas and brains of
REV is virus neutralization, although ELISA is the
REV‐infected chickens, but not from DNA from MD or
most  ­commonly used serological assay worldwide.
lymphoid leukosis lymphomas (5).
Immunoperoxidase plaque assay (35) was once shown to
Chronic neoplasia in the chicken where the tumors are
be a sensitive and reliable method for detection of REV
of bursal origin cannot easily be differentiated from lym-
antibody. Antibodies may be detected in serum or egg
phoid leukosis on pathologic criteria (251). Virological,
yolk from suspect birds by indirect immunofluorescence
serological, or PCR tests should be performed to confirm
(7, 257) or virus neutralization (148, 182). The agar gel
one oncogenic virus and exclude another.
precipitin test may detect viral antigen as well as anti-
Chronic neoplasia in the chicken in which bursal
body in sera (106, 107). Reticuloendotheliosis virus
tumors are lacking or in which the latent period is too
ELISA antibody kits are commercially available.
short for lymphoid leukosis must be differentiated from
Antibody tests are particularly useful in ascertaining lack
MD. Here too, pathological criteria are insufficient and
of exposure in specific pathogen free flocks or breeding
virological assays (including PCR) may be helpful. The
flocks producing progeny for export. Complementary
pp38 antigen of MD virus, occasionally expressed in MD
assays that are now very seldom used are reviewed in
lymphomas, is not present in RE lymphomas. Also, MHC
previous editions of this book.
class II (Ia) antigens are reported to be present on MD
lymphoma cells (199) but absent on RE nonbursal lym-
phoma cells (53). A comprehensive diagnostic strategy
Differential Diagnosis
for the differential diagnosis of virus‐induced lympho-
The pathology of REV‐induced lymphoproliferative mas in chickens has been introduced (253).
tumors can be confused with that of tumors seen in MD The acute reticulum cell neoplasia syndrome is not
and lymphoid leukosis (81, 238, 253). Thus, neoplastic known to occur in the field. A syndrome of broiler chickens
diseases must be diagnosed by exclusion; that is, by con- characterized by reticuloendothelial proliferation in the
firming specific agents involved and excluding other spleen and liver, and resulting in condemnation losses at
possible etiologies. Because avian tumor viruses are
­ processing, has been confused with RE (98, 239), but can
widespread and infection in the absence of tumor forma- be distinguished by the absence of RE antigens and proviral
tion is common, in many cases virological and serological DNA in the lesions. Virus detection tests including PCR
criteria do not always provide a definitive diagnosis. have been used to detect REV in a lymphosarcoma in an
However, diagnosis of RE should be supported by viro- Indian peafowl (152) and lymphomas in captive greater
logical evidence of REV infection, as REV is not as ubiq- and Attwater’s prairie chickens (74, 80, 268).
uitous as MDVs and ALVs. Techniques based on The runting disease syndrome must be distinguished
immunocytochemistry with mAbs to cellular, tumor, and from MD, especially when nerve lesions are present.
viral antigens, or molecular hybridization can be used in Differences between REV‐induced and MDV‐induced
the differential diagnosis of avian viral lymphomas includ- nerve lesions have been discussed (255, 257), but are not
ing RE, but are seldom used in diagnostic laboratories. consistent. Both types of nerve lesions must be distin-
Retroviral lymphomas in chickens originate from guished from spontaneous neuropathy, possibly an auto-
either B cells (RE, lymphoid leukosis) or T cells (RE), immune lesion of peripheral nerves (9).
whereas MD lymphomas are of T‐cell origin. The char- Lymphoproliferative disease (LPD) of turkeys can be
acteristics of target cells provide the basis for tests that confirmed or excluded by a combination of histopathol-
distinguish among B‐ and T‐cell lymphomas using mAbs ogy and molecular detection methods for REV and
specific for cell surface antigens of B‐ and T‐lympho- LPDV (1, 3). The PCR assays for LPD (1, 3, 197) and RE
cytes. Nondefective strains of REV have been shown to (5, 17, 38, 74, 78, 87, 92, 196, 268) should be useful, albeit
transform chicken B or T cells (109, 218). wild turkeys could bear dual infections with both viruses.
The PCR assays for RE, MD, and exogenous ALV can Marek’s disease has been diagnosed in turkeys in France,
be helpful in the differential diagnosis of RE. For instance, Israel, Germany, and Ukraine and should be ruled out in
because MD lymphomas should contain a significant the differential diagnosis.
proportion of MD virus‐infected cells, compared to In summary, naturally occurring RE lesions can be
latently infected tissues, MD lymphomas should have confused in the chicken with MD, LL, and various other
more infected cells, each with greater number of viral lymphoproliferative or immunodepressive conditions,
copies thus resulting in higher total estimates of viral and in the turkey, with LPD and MD.
Chapter 15  Neoplastic Diseases 637

Intervention Strategies Prevention and Control Procedures


Prevention of RE is currently accomplished through quality
Vaccination assurance of poultry biologics and, in SPF flocks, by strict
Reticuloendotheliosis in commercial poultry is typically biosecurity (250). It is desirable to prevent environmental
controlled by testing and elimination of infected shedder exposure and seroconversion of breeder flocks where prog-
breeding stock and thus vaccination is never considered. eny is destined for export, but this is difficult to accomplish
However, vaccines could be of use for immunization of because it may be impractical to truly prevent exposure in
endangered avian species. Vaccination of chickens with a the field. Control of insect vectors and fowl poxvirus infec-
recombinant fowl pox virus expressing the env gene of tion could be important in prevention programs (250).
REV (36, 167), or empty REV particles (151), provided Procedures for the control of RE have rarely been applied
some protection against REV infection. Defective REV in commercial practice, mainly because the disease has
particles (243) have been shown to induce neutralizing been sporadic and self‐limiting. Enzyme immunoassay to
antibodies (79). A baculovirus construct expressing the detect RE viral antigen in albumen samples seems to be the
env gene of REV has also induced REV antibody in chick- procedure of choice in commercial situations (108, 258).
ens (241). Presumably, it would be necessary to eliminate vertical
transmission through removal of potential transmitter
hens, and to rear progeny under isolated conditions whereby
horizontal infection could be precluded, as it has been done
Treatment
with ALV in chickens. Such control procedures could be
No treatment for RE is known. Since immune responses considered if REV infection becomes endemic in especially
are mounted to infection, it is possible that some affected valuable breeding stock, as is the case with the endangered
birds may recover. Attwater’s prairie chickens (27, 28, 74, 80, 87, 180, 250, 268).

­Other Tumors
Susan M. Williams, Rodney L. Reece, and Scott Hafner

Summary ­ heasants, and ratites) in which a known etiology does


p
not exist or is uncertain. Avian tumors often exhibit dis-
Agent, Infection, and  Disease.  Other tumors are in the tinctive microscopic appearances that may be difficult to
category where a known etiology does not exist or is extrapolate from similar mammalian tumors. The most
uncertain. The prevalence of these types of tumors will detailed examination of chicken tumors remains that of
depend on species, breed, age, and sex along with various Campbell (20) whose definition of a tumor as “an abnor-
intrinsic and extrinsic factors. mal tissue mass … which usually persists independent of
initiating factors … whose excessive, often uncoordi-
Diagnosis.  Avian tumors often exhibit distinctive micros­ nated growth threatens the host through compression,
copic appearances that may be difficult to extrapolate from infiltration, or remote spread” still suits our purposes.
similar mammalian tumors. Information on the classification Detailed information on the prevalence of different
and histologic appearance of some of the less common tumor types in poultry exists, but earliest reports are
tumors of poultry is available from detailed poultry skewed by a large number of virally‐induced “leukotic”
pathologist reports, zoo surveys, and noncommercial poultry tumors and the reproductive tumors that are common in
reports. Immunohistochemistry can aid in identification of adult hens. Tumor prevalence in poultry not only
tumor cells in poultry and other avian species with careful depends on species, breed, age, and sex, but also various
interpretation of the positive and negative controls. intrinsic (hormonal, genetic/developmental) and extrin-
sic (viral, chemical, and other environmental) factors
Intervention.  There is no intervention strategy due to (20). Some indications of the proportion of different
nature of these types of tumors. tumors to be expected in aged chickens unaffected by
exogenous viruses are available in the surveys of tumors
in specific pathogen free (SPF) flocks (38, 99). Information
Introduction on the classification and histologic appearance of some
of the less common tumors of poultry is available from
Other tumors refers to tumors of poultry (chickens, detailed reports by poultry pathologists, zoo surveys,
turkeys, quail, pigeons, ducks, geese, guinea fowl,
­ and examinations of the tumors of noncommercial birds.
638 Section II  Viral Diseases

Examinations of tumors present in poultry at slaughter yield insight into the development of this neoplasm in
are particularly useful as the sheer numbers of poultry both species (7, 13, 57, 60).
slaughtered may provide multiple examples of even very
rare tumors, and slaughtered birds are conveniently sep- Adenocarcinoma
arated by age and species. Radical deviations from the Early tumors are small, firm, white nodules that occur on
“normal” prevalence, distribution, and/or type of these the surface of the ovary; these may be mistaken for atretic
tumors may indicate changes in etiology or other factors follicles, but are less symmetric (37). Nodules coalesce
that require further elucidation. Immunohistochemical into a gray‐white, cauliflower‐like mass that commonly
techniques that utilize antibodies to lineage‐specific cell seeds serosal surfaces with transcoelomic metastases.
markers have found increasing applications as an aid to Ascitic fluid often develops with growth of the metasta-
identification of tumor cells in both poultry and other ses and terminally affected hens may assume an upright,
avian species. Extended formalin fixation may reduce the penguin‐like posture. Ovarian adenocarcinomas must
avidity of some antibodies for their designated tissue be differentiated from oviductal adenocarcinomas as
epitopes; this at times may be reversed by heated acid both may be widely metastatic and oviductal tumors may
solutions or applications of proteases. Some studies have metastasize to the ovary. Failure to detect tumors in the
utilized antibodies to vimentin for detecting mesenchy- lining of the oviduct suggests tumors are of ovarian
mal cells, cytokeratin for epithelial cells, actin for smooth rather than oviductal origin. Tumors are assumed to
and skeletal muscles, and neurofilament for nerves. arise from the mesothelial covering of the ovary (germi-
Some other antibodies may exhibit different avidity in nal epithelium), including its invaginations into the
avian tumors compared to that in mammalian species, ovary, but other suggested tissues include sex cord rem-
for instance S100 proteins may stain normal nerves, but nants, thecal cells, interstitial cells, or even the mesone-
not label tumor cells derived from those tissues (72). phros. Small tumors appear to be located in the theca
Neuron‐specific enolase often will intensely stain neural externa of small follicles (Figure 15.52) or in the ovarian
tissues, but may also stain other tissues, particularly stroma; a few may originate in the ovarian stalk. Multiple
muscle. Careful evaluation of the particular antibody in tumors may be present. Smaller tumors contain predom-
the both normal and neoplastic tissues of the affected inantly epithelial cells and little stroma, while larger
species including both positive and negative controls is tumors are often scirrhous. In advanced cases, ovarian
important for accurate interpretation of immunohisto- follicles fail to mature and oviducts are often atrophied.
chemical findings. Ovarian adenocarcinomas are not associated with
increased production of steroid hormones (37, 38).
Occasionally, ovaries affected by adenocarcinomas are
Public Health Significance covered with small cysts filled with yellow fluid; these
There is no known public health significance. may be dilated lymphatics or ovarian stromal lacunae
(83). A survey of 400 hens revealed approximate 2% inci-
dence rate of tumor restricted to ovary (60).
Histologically, ovarian adenocarcinomas are often
Urogenital System composed of acini lined by cuboidal to low columnar,
non‐ciliated, epithelial cells. These epithelial cells are
Ovary
often arranged around lumina that are at times filled
Despite the considerable differences between avian and with intensely eosinophilic periodic acid‐Schiff (PAS)
mammalian ovaries, particularly with regard to histo- positive material (Figure 15.53). In other, more densely
logic appearance and physiology, attempts are made to cellular tumors, acini may be compressed into cords or
characterize avian ovarian tumors using criteria devel- islands of epithelial cells or papillary proliferations of the
oped for mammals, particularly humans. Even with the lining epithelium may be invaginated into dilated acini
difficulty of categorizing avian neoplasms by mamma- (Figure 15.54). The mitotic rate is variable, and in many
lian criteria, classifying ovarian tumors as derived from cases mitoses are not prominent. In larger tumors, acini
surface mesothelium (adenocarcinomas), sex cord tis- are often surrounded by a marked scirrhous response
sues (granulosa cell tumors and arrhenoblastomas), (Figure 15.55), and in some serosal implants there may
germ cells (teratomas and dysgerminomas), or other be a proliferative response of the underlying muscularis
supportive tissues provides clues to both expected (84). Similar ovarian adenocarcinomas have been
behavior and possible etiology, in addition to differenti- described in turkey hens (119).
ating these tumors from metastatic disease. In particular,
ovarian adenocarcinomas of White Leghorn hens have Granulosa‐Theca Cell Tumor
recently been investigated as an animal model for ovar- Granulosa‐theca cell tumors are irregularly round to
ian cancer of women, with potential for this research to multilobular and encapsulated by a smooth, glistening
Chapter 15  Neoplastic Diseases 639

Figure 15.54  Ovarian adenocarcinoma with papillary structures


projecting into dilated acini. H&E, ×160.

Figure 15.52  Ovarian adenocarcinoma in the theca region


demonstrating delicate trabeculae and round nuclei; note the
granulosa cells and yolk of the developing ova (top). H&E, ×360.

Figure 15.55  Another form of ovarian adenocarcinoma with


dense bands of stromal cells enclosing clusters of neoplastic
acinar cells with intensely basophilic nuclei. H&E, ×160.

membrane. Tumors occasionally exhibit peripheral thin‐


walled cysts filled with fluid; sectioned tumors are yellow
and friable and some contain hemorrhages. These tumors
enlarge while remaining attached by the thin ovarian
pedicle. Tumors may become quite large without metas-
tasizing, but metastases to visceral organs and serosal
surfaces can occur (20, 37). Granulosa‐theca cell tumors
are by far the most common ovarian tumor of the young
Figure 15.53  Acinar structures, typical of ovarian
adenocarcinoma filled with eosinophilic material and lined by
broiler chicken. In these birds, there is often marked
cuboidal cells containing round nuclei with condensed chromatin precocious glandular hyperplasia of the oviduct (50).
­
and sparse eosinophilic cytoplasm. H&E, ×600. Histologically tumor cells are polyhedral to fusiform with
640 Section II  Viral Diseases

pale eosinophilic to vacuolated cytoplasm (Figure 15.56). that he designated a thecoma. A metastatic granulosa‐
Nuclei are generally irregularly round to ovoid. The theca cell tumor has been described in a duck (16). A very
arrangement of tumor cells varies widely from the forma- low incidence of  granulosa‐theca cell tumors has been
tion of follicular structures to solid sheets composed of induced in ­meat‐type chickens inoculated as embryos with
tightly packed thin cords (Figure  15.57) separated by a ALV‐J (92).
fine fibrovascular stroma, more elaborate gyriform
arrangements (Figure 15.58), or rosettes arranged around
small central spaces. The mitotic rate is variable but often
low, and the stroma may be prominent.
Ultrastructurally, tumor cells are identified by tran-
sosomes, a structure specific to avian granulosa cells and
those of some other lower vertebrates (37). Transosomes
(lining bodies) are dense structures of the lateral and apical
plasma membranes of granulosa cells that are taken up by
the oocyte becoming associated with primordial yolk gran-
ules. These structures are involved in the transport of vitel-
logenin (a yolk precursor) into the oocyte (76). Although
granulosa cells from normal avian follicles produce proges-
terone, theca interna cells produce testosterone, and theca
externa cells estrogen (94): hens with large granulosa‐theca
cell tumors exhibit markedly elevated plasma levels of
estrogen (37). Oviducts of affected hens are the same size Figure 15.57  Granulosa‐theca cell tumor composed of a uniform
as hens in lay, but eggs are not produced. A significant the- population of tightly packed tumor cells with plentiful, pale
cal cell component may be present in granulosa‐theca cell eosinophilic cytoplasm and uniform, round vesicular nuclei. Note
tumors; the presence of numerous vacuolated theca‐like the mitotic figure (arrow). H&E, ×600.
cells may lead to characterization of a granulosa cell tumor
as “luteinized” or even as a “luteoma” (20). Campbell (20)
also includes a tumor composed primarily of thecal cells

Figure 15.56  Lobules of vacuolated cells separated by moderate Figure 15.58  Gyriform arrangements of cells in one area of an
trabeculae in a granulosa‐theca cell tumor. H&E, ×200. ovary with a granulosa‐theca cell tumor. H&E, ×400.
Chapter 15  Neoplastic Diseases 641

Arrhenoblastoma (Arrhenoma) These tumors were composed of compact masses of


These tumors are associated with formation of seminifer- seminiferous tubules lying under the ovarian capsule.
ous tubules within the ovarian stroma and are accompanied The tubules were lined by a single, radially arranged layer
by evidence of sex reversal (virilism). Sex reversal in domes- of tall columnar Sertoli cells (Figure  15.60); variable
tic fowl was well recognized in ancient times. Most cases of numbers of interstitial cells were present between
sex reversal in poultry are due to destruction of the func- tubules. There was no obvious sex reversal or alteration
tional left ovary and formation of an ovotestis in the rem- in circulating hormones.
nants of the rudimentary right gonad rather than hormone
production by ovarian tumors (20, 36). In birds, the male is Dysgerminoma
the neutral sex and the young female is demasculinized by Dysgerminomas are the equivalent of mammalian ovar-
the production of her ovarian hormones (88). Well‐differ- ian seminomas, and have been detected in a few intersex
entiated arrhenoblastomas are histologically composed of (pseudohermaphroditic) pullets (123, 124). There was no
branching cords of columnar epithelium that are often two overt sex reversal, but there was masculinization of the
cells thick; these resemble primitive seminiferous tubules. comb and plumage of the head. Tumor cells were round
There is little, if any spermatogenesis. In less differentiated to polygonal and contained round to occasionally reni-
tumors, epithelial cells may be arranged in cords, nests, and form nuclei. Some tumors had metastasized to the liver
rosettes that form incomplete tubules (Figure 15.59) sepa- or peritoneum.
rated by a prominent interstitium that may contain intersti-
tial (Leydig) cells (98). Large tumors may be hemorrhagic.
Mesosalpinx
Arrhenoblastomas have been induced by the injection of
radioactive isotopes into the left ovary (121). Leiomyoma
Leiomyomas (at times termed fibroleiomyomas or even
Ovarian Sertoli Cell Tumors fibroids) are common in the ventral ligament of the ovi-
Ovarian Sertoli cell tumors in the chicken seem to be duct and the oviductal wall in domestic fowl, including
limited to the five cases described by Fredrickson (38). SPF hens (56). The prevalence of these tumors is varia-
ble, but may affect up to 60% of hens in the first egg lay-
ing cycle (6). Similar tumors were present in the oviduct
ligaments in some rapidly growing Japanese quail (35).
Leiomyomas of the ventral ligament of the oviduct are
usually solitary. Unless tumors are quite large, there is
little effect on oviductal function, but there may be
increased egg yolk peritonitis (77). In hens affected
by these tumors, there were elevated serum concentra-
tions of estradiol, and oviductal leiomyomas were
induced in hens treated with both diethylstilbestrol and

Figure 15.59  Arrhenoma from a hen that showed sex reversal. Figure 15.60  Ovarian Sertoli cell tumor composed of well‐defined
Network of epithelial cells arranged as ill‐defined cords and seminiferous‐like tubules lined by Sertoli cells. Stroma contains
tubules. H&E, ×350. (C.J Randall). interstitial cells. H&E, ×600.
642 Section II  Viral Diseases

progesterone (5, 6). Some reports (17) have also demon- mucosal dysplasia, typically discovered during close
strated that the tumor cells contain receptors for both examination of the mucosa of the oviduct (15).
estrogen and progesterone, suggesting steroid hormones Histologically, these nodules are well‐demarcated from
are likely to be involved in the etiology of these tumors. adjacent glands and are composed of concentrically
Many tumors are discrete, pale, solid masses less than arranged columnar epithelial cells with acidophilic cyto-
1 cm in diameter, but some may be several centimeters in plasmic secretory granules and pale enlarged nuclei
diameter and heavily vascularized (77, 98). Tumor cells (Figure 15.62). Adenocarcinomas are grossly evident as
are well‐differentiated, interlaced, smooth muscle cells small pink to grey, firm masses that protrude into the
bundled by variable amounts of connective tissue lumen of the oviduct and may invade through the mus-
(Figure 15.61). Mitotic figures are rare. These tumor cells cularis. There is often a distinct border between the nor-
are immunolabeled by α‐smooth muscle actin, desmin, mal magnal mucosa and tumor cells (Figure  15.63).
and vimentin (77). Metastases are often well‐encapsulated (Figure  15.64)
and may be composed of well‐differentiated cells.
Implants on the enteric serosa are often anaplastic cells
Oviduct
embedded in dense connective tissue (Figure  15.65)
Adenocarcinoma while implants on the oviduct serosa lack this intense
Adenocarcinomas originating in the oviduct most com- scirrhous response (Figure 15.66). The metastases of ovi-
monly arise in the upper magnum, with occasional ductal adenocarcinomas may in some cases be histologi-
tumors seen in the uterus (shell gland) or infundibulum. cally quite similar to those of ovarian adenocarcinomas
These tumors, like ovarian carcinomas, are often meta- and since the ovary is also a common site for metastasis,
static to the mesentery and serous surfaces of abdominal differentiating the tumors may be difficult. The ultras-
organs, especially to the pancreas and duodenum, but tructural characteristics of oviductal adenocarcinomas
also to the ovary and occasionally to the surface of the have been described (64). Immunohistochemical studies
liver and spleen. Tumor cells may be spread transcoe- have demonstrated that the cells of oviductal adenocar-
lomically in the ascitic fluid accumulating in the intesti- cinomas contain ovalbumin (59), but more recent stud-
nal peritoneal cavity. In contrast, metastases to the lung ies (42) have also identified ovalbumin in ovarian
may be hematogenous (2, 67). The prevalence of ovi-
ductal adenocarcinomas as determined by examining
the oviductal mucosa for tumors varied widely (5 to 81%)
in one survey (44) and in another survey only 4% were
restricted to the oviduct (60). In some studies (4), a posi-
tive correlation existed between egg weight and body
weight, and prevalence of tumors.
Oviductal adenocarcinomas in both chickens and tur-
keys may begin as small (2–10 mm) nodular areas of

Figure 15.62  Dysplastic adenomatous focus in a fold in the


Figure 15.61  Leiomyoma of mesosalpinx composed of smooth magnum showing clear demarcation from surrounding normal
muscle fibers arranged in compact whorls. Mitoses are absent from glands. The columnar epithelial cells are densely packed and
this field, and the nuclear/cytoplasmic ratio is low. H&E, ×600. oriented concentrically. H&E, ×400.
Chapter 15  Neoplastic Diseases 643

Figure 15.63  Magnal adenocarcinoma showing the well‐defined


margin between normal secretory tissue with cellular cytoplasm Figure 15.64  Implant of magnal adenocarcinoma deep in the
containing eosinophilic granules of ovalbumin (above) and very ovary showing capsule around adenocarcinomatous cells. Despite
lightly granular tumor cells (below). H&E, ×400. the apparent aggressiveness of this tumor, mitotic figures are not
prominent. H&E, ×200.

adenocarcinomas; the latter finding was attributed to


cellular dedifferentiation or return of the cell to a less dif- areas with solid sheets of cells. Nuclei are hyperchro-
ferentiated state. Cells of oviductal adenocarcinomas matic and there often is a high mitotic rate.
retain receptors for estrogen and progesterone (5) and
the growth of these cells in vitro is stimulated by estro- Seminoma
gen (4). In a recent study, few oviductal adenocarcino- Seminomas have been reported in chickens, ducks, quail,
mas displayed immunostaining for v‐erbB, the chicken guinea fowl, and pigeons (56). These tumors are com-
oncogenic form of the epidermal growth factor homolo- monly unilateral and consist of intratubular or broad
gous to human HER‐2/neu, but it was present in many sheets of round to polyhedral cells (Figure  15.68) with
ovarian adenocarcinomas, especially those which were eosinophilic cytoplasm and irregularly round, eccentri-
large (57). The organ or tissue of origin of many meta- cally placed nuclei. Multinucleated syncytia may be pre-
static adenocarcinomas observed in the abdomens of sent. Some tumors have been metastatic.
poultry cannot be determined and these are best
described as “of undetermined origin”. Interstitial (Leydig) Cell Tumors
Interstitial cell proliferations may be present in semino-
mas of chickens (20), but interstitial cell tumors have not
Testis
been described in poultry. Interstitial cells were a com-
Sertoli Cell Tumor ponent of a mixed cell tumor affecting both testes of a
Sertoli cell tumors are rare in chickens (20) and a few duck, but only the Sertoli cell component was present in
have been reported in Japanese quail (46), a duck (24), a metastatic site (73).
pigeon (96), and a goose (118). There is a single case
report of a tumor arising in the testicular remnants
Renal
of  an  incompletely surgically‐caponized chicken (111).
Elongated Sertoli cells are commonly intratubular Renal nephroblastomas and adenocarcinomas occur in
(Figure  15.67) and arranged at right angles to tubular chickens. Some are associated with infections with avian
basement membranes, while in other cases there may be leukosis/sarcoma viruses (see Leukosis/Sarcoma Group).
644 Section II  Viral Diseases

Figure 15.66  Magnal adenocarcinoma implanted on the serosa of


the isthmus is surrounded by little fibrous tissue. The dilated
acinar lumina are lined by cuboidal epithelium. H&E, ×200.

Figure 15.65  Compacted acini lined by cuboidal epithelium


surrounded by dense drifts of fibrous tissue in this cirrhotic implant
in duodenal serosa of a magnal adenocarcinoma. H&E, ×175.

Digestive System
Alimentary Tract
Squamous cell carcinomas have been most commonly
reported from the oropharynx, esophagus, and crop of
chickens (56). Both humans and chickens from certain
regions of northern China exhibit an increased preva-
lence of esophageal squamous cell carcinoma (108, 120),
suggesting that common etiologic factors could play a
role as chickens and humans may have common expo-
sures to drinking water, food, and other environmental
factors. However, detailed epidemiologic studies have
not identified a specific cause (120, 129). Such tumors
were often superficially ulcerated and composed of cords
and islands of anaplastic squamous epithelial cells, some Figure 15.67  Sertoli cell tumor in a quail. The tubule‐like
of which surrounded keratin (Figure 15.69). structures are lined by cells two layers deep. H&E, ×360.
Chapter 15  Neoplastic Diseases 645

Figure 15.70  Adenocarcinoma of gizzard with growth of darkly


staining cuboidal tumor cells downward into muscularis. The
keratinous product of these cells is shown at lower right. H&E,
×200. (K. Langheinrich)

Adenocarcinomas of the small intestine have been


described in chickens (56) and ducks (104); some of these
involved the ileocecal junction. Primary adenocarcino-
mas affecting the intestinal tract must be differentiated
from metastatic adenocarcinomas of the reproductive
Figure 15.68  Seminoma in a duck. Lobules of pleomorphic tract, which readily and rapidly metastasize to involve
polyhedral cells with finely granular cytoplasm; some
multinucleated cells. Delicate stroma. H&E, ×180.
the intestine (20).
Enterogenous cysts that are lined by mucosa derived
from the gastrointestinal tract have been described in
young chickens. These usually present as fluid‐filled
cysts partially replacing the spleen and the lining mucosa
often resembles that of the ventriculus (69).
Leiomyomas and leiomyosarcomas have been described
affecting the gastrointestinal tract (see Musculoskeletal
System). Pheasants are affected by a nodular typhlitis
caused by infection with Heterakis isolonche with larval
migration producing neoplastic submucosal nodules that
are neurofibroblastic (63) or smooth muscle in origin (79)
with possible metastasis to the liver.

Liver
Hepatocellular Tumors
Figure 15.69  Oropharyngeal squamous cell carcinoma in an adult
noncommercial chicken. Cords and islands of epithelial cells with
Hepatocellular adenomas (hepatomas) and hepatocellular
some central keratin pearls (arrow). Mucosal gland is shown at top carcinomas are uncommonly reported in chickens (56).
left. H&E, ×200. Adenomas consist of masses of large, polyclonal, well‐dif-
ferentiated hepatocytes that are compactly arranged in
Multiple papillomas have been described in the esoph- several cell thick cords separated by connective tissue septa
agus and crop of chickens, but that report lacks histo- (Figure 15.71); portal triads and central veins are, however,
logical characterization (87). lacking in these masses and there is a low mitotic rate.
In chickens, proventricular adenomas (21) and adeno- Hepatocellular carcinomas often multifocally replace the
carcinomas (20, 99) have been described, along with liver parenchyma and there may be metastasis to the lung.
adenocarcinomas (20, 21, 98) of the gizzard (Figure 15.70). In carcinomas, the tumor cells are less well differentiated,
A proventricular adenoma was described in a duck (10). often exhibit mitoses, and may be multinucleated (20, 95).
646 Section II  Viral Diseases

Hepatocellular tumors in chickens have been induced with Peritoneum


some oncogenic avian retroviruses (14) or by administra-
Mesothelioma
tion of diethyl‐nitrosamine (68). These tumors appear to
Mesotheliomas have been reported in chickens (56) and
be not uncommon in ducks (15, 104) and some have been
ducks (74). Multiple papilliform abdominal nodules in
induced by feeding aflatoxin (23). Duck hepatitis B virus
which mesothelial cells and anaplastic fibrocytes were
has been suggested to be involved in the development of
seen have been described in an aged female ostrich (87).
hepatocellular carcinomas in Chinese ducks (31, 127), but
There often is ascites, and tumors invest serosal surfaces
aflatoxins were also suspected.
of abdominal organs. Histologically, papillae are covered
by cuboidal mesothelial cells supported on delicate stalks
Cholangiocellular Tumors
(Figure 15.76).
Cholangiomas and cholangiocellular carcinomas are
uncommon in poultry, with only a few cases being described
in chickens, ducks, and pigeons (56). Tumors of bile ducts
must be differentiated from the proliferation that accompa-
nies chronic toxic damage to the liver due to the ingestion of
hepatotoxins. Cholangiomas consist of clusters of dilated
and often distorted ducts that are lined by well‐differenti-
ated epithelial cells; these ducts are separated by fibrous
connective tissue (Figure  15.72). In cholangiocarcinomas,
epithelial cells less commonly form defined ducts and are
separated by fibroblasts (Figure 15.73).

Pancreas
Adenocarcinoma
Tumors of pancreatic epithelial cells are often difficult to
distinguish from metastases of ovarian or oviductal
tumors, as those tumors commonly implant upon the
duodenal serosa and surface of the pancreas. Pancreatic
tumors may arise from ducts (86, 97) or exocrine cells
(38). Most probably arise from ductal epithelium and are
composed of columnar epithelial cells with lightly Figure 15.72  Cholangioma composed of dilated ducts in a loose
basophilic cytoplasm (Figure  15.74). Tumors derived
­ fibrocytic stroma. H&E, ×75.
from  exocrine cells morphologically resemble acini
(Figure  15.75) and may or may not have cytoplasmic
bodies resembling zymogen granules (38).

Figure 15.71  Hepatoma composed of large eosinophilic neoplastic Figure 15.73  Cholangiocarcinoma composed of small clusters of
cells, some in mitosis, forming irregular plates. H&E, ×600. epithelial cells in a fibroblastic stroma. H&E, ×190.
Chapter 15  Neoplastic Diseases 647

Figure 15.76  Mesothelioma with prominent neoplastic epithelial


cells supported on a delicate stalk. H&E, ×600.

Figure 15.74  Pancreatic adenocarcinoma, probably of ductule


cell origin, composed of columnar cells forming tubular structures
among a few remnant acinar cells (arrow) ×160.

Figure 15.77  Adenocarcinoma of lung composed of a papillary


growth of epithelial cells that have replaced most of the normal
lung. H&E, ×200.

Lung
Adenocarcinoma
In chickens, primary adenocarcinomas of the lung
Figure 15.75  Pancreatic acinar cell adenocarcinoma with
appear to be extremely rare (8, 20). Campbell described
normal exocrine tissue (right) and agranular neoplastic cells (left).
H&E, ×600. three cases characterized by small nodules located near
the primary bronchi and Fredrickson and Helmboldt
described a papillary adenocarcinoma arising multifo-
cally from parabronchi (38). In that case, cuboidal
Respiratory System ­epithelial cells replaced most of the lung (Figure 15.77)
and there were metastases in the thorax and abdomen.
Infraorbital Sinus
Primary tumors of the lung, especially in mature
Reece (98) reported two chickens with pea‐sized cystic ­chickens, need to be ­differentiated from metastases of
adenomas that extended into the infraorbital sinuses and ovarian or oviductal adenocarcinomas, hepatocellular
were filled with mucin. carcinomas, or ­ metastases of other tumors (20, 99).
648 Section II  Viral Diseases

Reports of pulmonary adenocarcinomas are more


­common in ducks (75, 113, 128), particularly those main-
tained in zoos, than in other avian species. There are rare
descriptions in pigeons (113).

Nervous System
Central Nervous System
Astrocytoma
Both sporadic and epizootic cases of astrocytoma (fowl
glioma) have been described, commonly as multiple
tumors in aged hens (56), and one case has been
described in a domestic duck (110). These small masses
are often near the thalamus or the base of the cerebellum
(38). There frequently are perivascular accumulations of
lymphocytes adjacent to the tumor, but no hemorrhage,
Figure 15.79  Astrocytoma composed of uniform astrocytes with
giant cells, or areas of pressure necrosis (Figure 15.78). extended cytoplasmic processes. H&E, ×190.
Masses are composed of astrocytes with extended cyto-
plasmic fibrillar processes (Figure 15.79). Similar masses

in some breeds of chickens have been shown to be caused


by infections with a specific subgroup A avian leukosis
virus (61, 85, 116).
The small ependymoma described by Wight and
Campbell (125) was discovered in the right cerebral
hemisphere of a 60‐day‐old chicken affected by MD;
the mass was within or adjacent to the lateral ventricle
and consisted of vacuolated cells that occasionally
formed palisades or rosettes. In the same report, two
subdural angioblastic meningiomas were described in
pullets displaying neurological signs: both were located
near the cerebellum and consisted of multiple con-
gested vascular sinuses that were lined by enlarged
endothelial cells and separated by a prominent reticulin
network.

Pineal Body Tumor


There are a few reports of pineal body tumors in chick-
ens (20, 99, 115). In the young hen described by Swayne
et al. (115), the mass was somewhat similar to a normal
pineal gland, but was greatly enlarged (3×), displaced
adjacent cerebellar tissue, and cells exhibited an
increased mitotic rate. These characteristics, in addi-
tion to a relatively decreased number of follicular cells,
identified this as a tumor rather than pineal gland
hyperplasia. In the cases reported by Reece (99), there
were clinical histories that included neurologic symp-
toms such as fine tremors and head pressing. In each
bird, there was a large mass between the cerebrum and
cerebellum that extended into or impinged upon the
Figure 15.78  One of several clearly demarcated, but cerebellum. Tumors were composed of lobular masses
unencapsulated astrocytomas composed of fibrillar astrocytes, in
of low columnar epithelial cells (Figure  15.80) sur-
anterior brain stem. There is a significant lymphocytic infiltrate
around the blood vessels within the tumor and the adjacent rounding a central lumen. These cells were surrounded
tissue. H&E, ×100. by parafollicular cells.
Chapter 15  Neoplastic Diseases 649

Figure 15.81  Schwannoma of the sciatic plexus showing a whorling


pattern of spindle‐shaped cells with central nuclei. H&E, ×400.

Figure 15.80  Lobules of a pineal body tumor separated by fine


then entrapped within, densely collagenous scar tissue,
trabeculae. Palisaded low columnar epithelial cells with large vesicular
nuclei are surrounded by smaller parafollicular cells. H&E, ×200. resulting in tangled masses of poorly myelinated axons,
Schwann cells, and perineurial cells separated by bands of
connective tissue (Figure 15.82). These neuromas are not
Peripheral Nervous System
true neoplasms and are a well‐recognized although rela-
Neurofibromas tively rare, sequela to beak‐trimming (Figure 15.83) or toe
The term neurofibroma is often used interchangeably with amputation (39, 40). When young chicks or turkey poults
Schwannoma (neurilemmoma), however, Schwannomas are partially beak‐trimmed, the resultant dermal scar tis-
are relatively homogenous proliferations of Schwann sue is less dense which may p ­ artially explain the relative
cells, while neurofibromas contain a mixture of nerve absence of neuroma development in birds treated when
elements including Schwann cells. Separation of these they were young (28, 41).
tumors is not always possible in poultry and similar
tumors have been referred to by some authors as neuro-
Melanoma
genic sarcomas (87). Tumors of the nerve sheath are well
recognized in young broiler chickens (21) but also occur Melanomas, including some described as originating in
in older birds, often originating in major nerves or near the ovary with metastasis to other organs have been
dorsal root ganglia. Neurofibromas are often solitary described in the chicken (20). Multifocal melanomas with
masses, but multiple tumors have been described (3). the appearance of malignancy have been seen affecting
Tumors are composed of fibroblast‐like spindle cells that the skin, visceral organs, musculature, and other tissues in
may form concentric whorls resembling nerve sheaths young broiler chickens and a 16‐week‐old pullet (99).
(Figure 15.81). In tumors with ­predominantly Schwann Some authors described similar tumors as malignant mel-
cell proliferation, there may be nuclear palisading or anomas and demonstrated that unbleached tumor cells
structures resembling sensory nerve endings (Wagner– and tumor cells bleached by potassium permanganate or
Meissner corpuscles or Pacinian corpuscles) (20). hydrogen peroxide, were not immunoreactive with
­antibodies specific for S100 proteins, but did react with
Neuromas antibodies to neuron specific enolase, vimentin, and
Post‐traumatic neuromas commonly occur when afferent Melan‐A (126). Melanocytes in avian melanocytic tumors
nerves attempting to regenerate become obstructed by, often do not react with antibodies to S100 proteins even
650 Section II  Viral Diseases

Figure 15.83  Lower beak neuroma due to beak trimming. The mass


can range in size and may hinder food intake if large enough.

Figure 15.82  Post‐traumatic neuroma in beak tip showing


dense collagenous scar tissue and multiple whorls of nervous
tissue composed of poorly myelinated axons, Schwann cells,
and associated connective tissue. Martius scarlet blue, ×360.
(C.J Randall)

though normal nerves are S100 immunopositive (55, 72).


These cells may be spindloid or there may be closely
packed masses of polyhedral melanocytes reminiscent of
epithelial tissue (Figure  15.84). Malignant melanomas
have also been described in ducks and a pheasant and
melanomas, some of which were amelanotic, have been Figure 15.84  Subcutaneous melanoma of the wing of a pigeon.
seen in the skin of pigeons (56). Benign melanomas have There is melanin pigment in many polyhedral cells. Mitotic figures
are rare. The cells are densely packed, clearly demarcated from the
been seen within the eye of chickens (12, 32, 107). A small surrounding tissue, and penetrate between muscle fibers and
number of cases of multifocal melanomas were noted in adipocytes (lower right). H&E, ×200.
young adult Japanese quail from one inbreed commercial
strain (101).
Endocrine System
Adrenal Gland
Special Senses
The avian adrenal gland is composed of intermingled
cords of cortical cells and enterochromaffin (medullary)
Eye
cells rather than displaying a defined cortex and medulla.
Intraorbital rhabdomyosarcomas, thought to originate Two adrenal cortical adenomas have been described, one
from the striated muscle of the iris, have been described in a young chicken and one in an SPF hen (21, 99). The
in chickens (32). Retinoblastomas (20, 26) and melano- well‐differentiated cells comprising these neoplasms
mas involving the eye, have been described. contained abundant eosinophilic cytoplasm that was
Chapter 15  Neoplastic Diseases 651

occasionally vacuolated. The close anatomic relationship Hemangiopericytoma


between the ovary and adrenal glands makes it impera- A few hemangiopericytomas have been described in
tive to eliminate markedly luteinized ovarian ­carcinomas chickens (38, 112), they were all benign and occurred
when the ovary is also involved (20). A pheochromocy- as  subcutaneous nodules in the cervical region.
toma has been described in a 14‐week‐old pullet (20). Histologically, they were arranged as concentric rings
around blood vessels, and were composed of uniform
spindle‐shaped cells with a fusiform nucleus. The inter-
Pituitary Gland vening reticulin fibers could be demonstrated by a silver
Pituitary tumors of poultry appear to be limited to the stain (Figure 15.85).
two chickens reported by Campbell (20). One was
described as a tumor of chromophobe cells that com- Lipoma and Liposarcoma
pressed the brain stem, and the second was an infiltrative Subcutaneous lipomas are not common in chickens (20,
acidophil adenoma. 97). They are generally benign, encapsulated and deli-
cately trabeculated; and may exhibit necrosis and/or
hemorrhage. These tumors are composed of large mature
Thymus
vacuolated adipocytes with a pale displaced nucleus:
Thymomas have been most commonly described in mitotic figures are rare. Liposarcomas of chickens are
chickens and one has been described in a duck (56). These rare and may be locally invasive or metastasize (100); in
have primarily been masses in the neck that are histologi- some cases, the predominant cells are elongate and may
cally characterized by sheets of epithelial cells lacking be ­mistaken for fibrocytes except for the presence of
­distinct cytoplasmic borders. Immunostaining of tumor small cytoplasmic fat vacuoles, others are composed of
cells with cytokeratin has been utilized to verify epithelial more typical immature adipocytes. Myelolipomas and
origin (11). There may be accompanying myoid cells (20). erythrolipomas have not been reported thus far in
chickens.

Thyroid and Parathyroid Glands


Adenomas of the thyroid have been described in a 24‐
week‐old pullet (87) and a 2‐year‐old hen (20). Both
affected the left thyroid glands, contained multiple cysts
and were described as containing relatively undifferenti-
ated epithelium resembling embryonic thyroid.
Microscopic cysts lined by ciliated epithelium are occa-
sionally seen in the thyroid and are thought to be derived
from thyroglossal duct remnants (20). Tumors of the
parathyroid gland of chickens seem to be limited to an
adenoma and a parathyroid carcinoma (48).

Integument
Subcutis
Of the various sarcomas, fibromas, and myxomas that are
encountered in subcutaneous tissues of chickens, many
may be induced by avian leukosis viruses (see Leukosis/
Sarcoma Group), although some of these tumors are
reported from SPF hens and other poultry species.
Chickens systemically infected by some retroviruses may
produce tumors in response to the inflammation
­associated with wounds, as postulated to be the cause of
some mesenchymal tumors observed near the trimmed
beak of hens (99). There is a report of a fibroma in a wild,
hunter‐killed, ring‐necked duck with a large mass on the
side of the head from the commissure of the beak to ven- Figure 15.85  Hemangiopericytoma with concentric rings of
tral of the ear (19). pericytes clearly defined. Silver, ×90.
652 Section II  Viral Diseases

Cutis i­dentified (109), but in other studies, viral particles were


not seen ultrastructurally and evidence of avian leukosis
Squamous Cell Carcinoma
viruses was not detected (29, 52, 81). Carcasses most
The true squamous cell carcinoma of the skin of birds
commonly exhibit crater‐shaped ulcers with raised
originates from the surface epidermis, forming infiltrat-
­margins within feather tracts (Figure  15.86), but both
ing cords of anaplastic keratinocytes that invade down-
nodular and ulcerative lesions may be present. Smaller
ward into the underlying dermis. Epithelial cells are
circular ulcers average 5 mm in diameter, but larger
separated by intercellular bridges and resemble cells of
irregular, coalescing ulcers are present on some ­carcasses.
the stratum spinosum. Since it is likely that some of these
In live chickens, these ulcers are filled with keratin and
tumors, like those of other species, are induced by years
cell debris (Figure 15.87). Nodules are generally smaller
of sun exposure, many have occurred on the feet and
(averaging 3 mm in one study) and appear grossly as
shanks of aged birds (20, 22). Although these tumors are
locally invasive, only a few have been metastatic (1). The
tumors originally described as dermal squamous cell
carcinomas in young broiler chickens, have been desig-
nated avian keratoacanthomas, due in part due to origin
from feather follicle epithelium rather than surface epi-
dermis (109). Oropharyngeal, esophageal, and ingluvial
squamous cell carcinomas also have been recognized,
especially in chickens (see Digestive System).

Avian Keratoacanthoma
These tumors are most commonly found in the skin of
carcasses of young chickens at slaughter (56), although
some have been identified in live broiler chickens (52, 99)
and a few have been seen in older chickens (66, 114).
These tumors were formerly designated as “dermal squa-
mous cell carcinoma”, but avian keratoacanthoma was
selected as a more descriptive term since tumors unal-
tered by the processing artifacts of de‐feathering and
Figure 15.86  Typical carcass lesions of avian keratoacanthomas
scalding have the distinctive tissue architecture of kera- are craterous ulcers within feather tracts.
toacanthomas, originate from feather follicle epithelium
rather than the surface epidermis, are not metastatic,
and, in live birds, regress (52). Carcasses with extensive
lesions are condemned at slaughter, while less affected
carcasses undergo trimming. The prevalence of broiler
carcasses with multiple lesions commonly varies from
0.01% to 0.05%, but may be 0.09% or higher in individual
flocks (53, 117, 122). In some studies, chickens slaugh-
tered at less than 48 days of age exhibited more tumors
than older birds and, tumor prevalence was cyclic, being
lowest in summer months (53, 122). In other surveys, an
increased number of condemnations due to these tumors
were associated with dusty houses, birds placed in new
houses, or certain farms (43). One report has commer-
cial adult layer hens (55,000 hens) affected starting at 30
weeks of age and continuing for 28 weeks and regressing.
Approximately 0.1% of the flock was affected in the
region of the legs and toes (29).
The etiology of this condition remains unknown. Some
authors have suggested an association with fowl pox
because some studies by nested PCR detected DNA
sequences specific for fowlpox virus in lesions (33), but Figure 15.87  In live chickens with keratoacanthomas, ulcerative
other studies failed to detect evidence of fowlpox (81). In lesions contain central masses that are mixtures of keratin, cell
one study of these lesions, type C retroviruses were debris, and bacteria.
Chapter 15  Neoplastic Diseases 653

enlarged feather follicles (Figure 15.88). Microscopically, Ulcers are composed of a central cup‐shaped cavity lined
nodular lesions appear as proliferative outgrowths of by epithelium and filled with keratin, bacteria, and cell
feather follicle epithelium (Figure 15.89), cysts originat- debris. Epithelial lips overhang this central mass of kera-
ing from dysplastic feather follicles, or hyperplastic tin and cell debris. The lining epithelium keratinizes
feather follicles that contain hyperkeratotic feathers. toward the central cavity and extends thin strands of
keratinocytes into the surrounding dermal fibroplasia
(Figure  15.90). Carcass lesions are often extensively
altered by post‐slaughter de‐feathering with loss of the
central keratin core and much of the lining epithelium. In
affected live chickens, nodules progressed to ulcers and
all lesions eventually regressed (52).

Feather Folliculoma
Multiple cystic structures found on the medial surfaces
of the wings of adult hens have been described (99).
These cysts were filled with keratin and feather rem-
nants, and were lined by squamous epithelial cells. In
some areas, there was abrupt keratinization of the lining
epithelium, while in other areas, there was disorganized
feather follicle epithelium (Figure  15.91). There was an
intense inflammatory reaction in the adjacent dermis.
Similar cysts have been described in turkeys (27).

Intracutaneous Keratinizing Epithelioma


These benign masses were characterized by multiple
nodules in the facial skin of adult hens, each marked by a
central pore. These cysts were lined by stratified squa-
mous epithelium with orderly maturation from a periph-
Figure 15.88  Early keratoacanthoma lesions in the skin of live eral basal layer. The cyst lumen contained lamellated
chickens are nodules in the base of feather follicles.
keratin (Figure  15.92) but no feather remnants. Little

Figure 15.90  A section through the epithelial lip of an ulcerative


Figure 15.89  Microscopically, nodular lesions of avian keratoacanthoma from a live chicken shows the centrally
keratoacanthoma are expansions of feather follicle epithelium. keratinizing and peripherally invasive lining epithelium (arrows)
H&E, ×40. adjacent to the central keratin and cell debris. H&E, ×100.
654 Section II  Viral Diseases

Figure 15.92  The lumen of this intracutaneous keratinizing


Figure 15.91  Edge of a feather folliculoma showing dysplastic epithelioma contains lamellated keratin. The epithelium shows basal
specialized feather‐forming epithelium and an adjacent cord of cells aligned on a distinct basal lamina and progression to polyhedral
basal cells. The lumen was lined by stratified cuboidal to cells with abrupt keratinization. Note the small intraepithelial bulla.
squamous epithelium with abrupt keratinization and contained H&E, ×190. (Reece (99). Courtesy of Taylor and Francis, Ltd, www.
keratin and feather remnants. H&E, ×180. tandfonline.com, on behalf of Houghton Trust, Ltd.).

surround cholesterol clefts. The multiple xanthomas


inflammation surrounded these cysts unless there was seen in chickens in the 1950s were attributed to chlorin-
rupture of the cyst wall (99). ated hydrocarbons contaminating the fat added to
rations. The masses were considered to possibly be a
Other Tumors of the Cutis reaction to metabolites of this material accumulating in
Acanthomas are firm, often conical masses that pro- dermal fat (106).
trude from the scaled epithelium on the plantar surface
of the tarsometatarsus. Tumors are composed of keratin
whorls on a base of fibrous connective tissue that con-
tains islets of squamous epithelium with central basal
Musculoskeletal
epithelial cells (20).
Leiomyoma and Leiomyosarcoma
Mast cell tumors are extremely rare in poultry, but
have been described in a few adult chickens (54, 58, 91). Leiomyomas of the ventral ligament of the oviduct are
In one, there was a solitary lesion of the eyelid, while in common in domestic fowl and Japanese quail (see
the other two birds, there were multiple dermal tumors Urogenital System), other smooth muscle tumors are
composed of solid sheets of round to ovoid mast cells. In more rarely reported. Leiomyomas have been described
one hen, there was metastasis to the lung (54). in the intestine of ducks (98), the trachea of broiler
Xanthomas are not true neoplasms, but are masses of ­chickens (21, 98, 102) (Figure  15.93), attached to the
foamy macrophages and multinucleated giant cells that ­pancreas (97) or in the liver of pigeons (25), and in the
Chapter 15  Neoplastic Diseases 655

Figure 15.93  Leiomyoma of the trachea of a broiler chicken.


Bundles and whorls of smooth muscle fibers in the lamina propria
(upper portion) penetrate between the cartilaginous rings and
extend into the adjacent adventitia (lower right). H&E, ×100.

muscularis of chicken proventriculus, ventriculus (giz- Figure 15.94  Rhabdomyosarcoma. Some cells are strap‐like,
whereas others are large and polyhedral. The cytoplasm is
zard), and duodenum (21, 99). Leiomyosarcomas have eosinophilic and some cells contain multiple nuclei. H&E, ×400.
been described in chicken tracheal muscle (20), chicken
ventriculus (gizzard) (105), hen ovary (65), chicken
intestine (3), and thigh skeletal muscle of a hen with
metastasis (70). Leiomyosarcomas have also been
described in the lung and skin of pigeons (80, 82).

Rhabdomyoma and Rhabdomyosarcoma
Rhabdomyomas and rhabdomyosarcomas have been pri-
marily seen affecting the skeletal muscle of the breast and
legs and less commonly the musculature of the hearts of
young chickens (20, 21, 87), and a rhabdomyoma has also
been described in the eyelid of a racing pigeon (97). A
rhabdomyosarcoma originating in cranial muscle and
invading the brain occurred in a 7‐month old hen (47),
and rhabdomyosarcomas metastatic to the lung of chick-
ens have also been described (71, 99). Two intraocular
rhabdomyosarcomas have been described in chickens
(32). These muscle cell tumors are generally histologically
characterized by spindle, strap, flame, or racquet cells and
multinucleated cells (Figure 15.94). Cross striations may
be present only in rare cells; even examinations with
polarized light or phosphotungstic acid‐stained sections Figure 15.95  Osteoma showing thick irregular trabeculae. H&E, ×100.
may not detect striations in the intensely eosinophilic
cytoplasm of anaplastic myoblasts. Immunohistochemical
examinations for vimentin, myoglobin, muscle specific These osteomas were well circumscribed and were com-
actin, and desmin have been useful in some avian posed of disorganized bone trabeculae (Figure  15.95).
rhabdomyosarcomas (34). Several osteosarcomas, some of which were metastatic,
were described affecting long bones, ribs, and vertebrae
of young chickens (21). Another osteosarcoma affecting
Osteoma and Osteosarcoma
an aged hen was also metastatic (110). A report describ-
Osteomas have been described in the feet of ducks (97) ing osteosarcoma in free range aged hen had involve-
and affecting various sites in a few chickens (20, 21, 99). ment of the vertebrae with metastasis to the liver (30).
656 Section II  Viral Diseases

An osteosarcoma has also been described affecting the that surround keratin (resembling primitive feather fol-
tibiotarsus of a Japanese quail (97) and the foot of a licles). Some tumors are described simply as abdominal
goose (78). masses (62). In chickens, teratomas are reported to arise
more commonly in the testis than ovary (20, 21, 62).
Teratomas may also arise at other sites (20, 21, 48, 93).
Chondroma and Chondrosarcomas
Teratomas have also been induced in chickens by the
A single chondroma has been described as originating at injection of various metallic ions into the testis (20, 49).
a costochondral junction in a young chicken (20). Teratomas have been observed in several ducks and a
Multifocal chondromas have been described in the foot- goose (18, 89, 103).
pads of geese and ducks (97). These tumors were charac- In contrast to teratomas, hamartomas are a focal over-
terized by lobules of chondrocytes separated by trabeculae growth of mature tissue indigenous to the organ or
(Figure 15.96). Dittmer et al. described a chondrosarcoma ­location in which it is found; these have not been reported
in an aged free range hen that involved the sternum and in poultry.
extended into muscle (30). Chondrosarcomas in poultry
are exceedingly rare.
Multicentric Histiocytosis
Multicentric histiocytosis (histiocytic sarcomatosis, sys-
Other Tumors temic spindle‐cell proliferative disease) primarily affects
young broiler chickens, producing hepatomegaly and
Teratoma splenomegaly. Numerous small (0.5 to 2 mm) white
masses are grossly evident in the spleen, liver, and kid-
Teratomas are composed of tissues arising from more neys. Some diseased birds are pale (anemic) and smaller
than one germinative layer and commonly contain a dis- than flock mates. Microscopically, nodules of spindle‐
organized mixture of variously differentiated epithelial shaped cells replace the spleen (Figure 15.97), liver, kid-
cells, bone, cartilage, smooth muscle, fat, or other tissues neys, and other organs most commonly including bone
including nervous tissue, melanocytes, or cardiac mus- marrow, pancreas, intestine, proventriculus, and lungs
cle. Often, ciliated columnar epithelial cells with goblet (9, 45, 51, 90, 91). Germinal centers may be present
cell differentiation line tubules or cysts. Other epithelial within these nodules, especially in the spleen; liver nod-
islands may be composed of squamous epithelial cells ules may contain a more heterogenous population of
cells, including plasma cells and some typically bulge
into portal vessels. Lesions are not accompanied by mye-
loid leukosis (myelocytomatosis). Spindle cells contain
abundant eosinophilic cytoplasm and elongated ovoid to
more pleomorphic nuclei (Figure  15.98). Mitoses are

Figure 15.96  Multifocal chondroma of the footpad of a goose


showing lobules of cartilage that are separated by fibrovascular Figure 15.97  In multicentric histiocytosis, nodular masses of
trabeculae. H&E ×100. spindle‐shaped cells multifocally replace the spleen. H&E, ×100.
Chapter 15  Neoplastic Diseases 657

common, but there are no multinucleated cells. Spindle


cells are immunohistochemically positive for markers
characteristic of antigen‐presenting tissue macrophages
or dendritic cells (9, 90). Infection of meat‐type chickens
at day of hatch with strains of subgroup J avian leukosis
virus have reproduced these lesions, but only in birds
that are persistently viremic with an ineffective antibody
response (90).

­Acknowledgement
The authors would like to acknowledge the contribu-
tions of Drs. Bruce W. Calnek, Aly M. Fadly, Karel A.
Schat, and Richard L. Witter for their contributions
to  subchapters in Neoplastic Diseases in previous
Figure 15.98  Histiocytic spindle‐shaped cells in the spleen of a editions.
chicken with multicentric histiocytosis contain elongated and
pleomorphic nuclei. H&E, ×400.

­References

Introduction Research, K.A. Schat, et al., eds. American Association


of Avian Pathologists, Kennett Square, PA. 1–9.
1 Javier, R.T., and J.S. Butel. 2008. The history of tumor
10 Read, A.F., S.J. Baigent, C. Powers, L.B. Kgosana, L.
virology. Cancer Res. 68:7693–7706.
Blackwell, L.P. Smith, D.A. Kennedy, S.W. Walkden‐
2 Calnek, B.W. 1992. Gordon Memorial Lecture. Chicken
Brown, and V.K. Nair. 2015. Imperfect vaccination can
neoplasia—a model for cancer research. Br Poult Sci.
enhance the transmission of highly virulent pathogens.
33:3–16.
PLoS Biol. 13: e1002198.
3 Osterrieder, N., J.P. Kamil, D. Schumacher, B.K. Tischer,
11 Davidson, I., M. Malkinson, and Y. Weisman. 2002.
and S. Trapp. 2006. Marek’s disease virus: from miasma
Marek’s disease in turkeys. I. A seven‐year survey of
to model. Nat Rev Micro. 4:283–294.
commercial flocks and experimental infection using
4 Parcells, M., J. Burnside, and R. Morgan. 2012. Marek’s
two field isolates. Avian Dis. 46:314–321.
disease virus‐induced T‐cell lymphomas. In: Cancer
12 Murata, S., K.S. Chang, Y. Yamamoto, T. Okada, S.I. Lee,
Associated Viruses‐Current Research, E.S. Robertson,
S. Konnai, M. Onuma, Y. Osa, M. Asakawa, and K.
ed. Springer Science+Business Media. 307–335.
Ohashi. 2007. Detection of the virulent Marek’s disease
5 Zhao, Y., H. Xu, Y. Yao, L.P. Smith, L. Kgosana, J. Green, L.
virus genome from feather tips of wild geese in Japan and
Petherbridge, S.J. Baigent, and V. Nair. 2011. Critical role of
the Far East region of Russia. Arch Virol. 152:1523–1526.
the virus‐encoded microRNA‐155 ortholog in the induction
13 Witter, R.L., B.W. Calnek, C. Buscaglia, I.M. Gimeno,
of Marek’s disease lymphomas. PLoS Pathog. 7: e1001305.
and K.A. Schat. 2005. Classification of Marek’s disease
6 Kaufer, B.B., S. Arndt, S. Trapp, N. Osterrieder, and K.W.
viruses according to pathotype—philosophy and
Jarosinski. 2011. Herpesvirus telomerase RNA (vTR)
methodology. Avian Pathol. 34:75–90.
with a mutated template sequence abrogates
14 Gimeno, I.M., R.L. Witter, A.L. Cortes, and W.M. Reed.
herpesvirus‐induced lymphomagenesis. PLoS Pathog. 7:
2011. Replication ability of three highly protective
e1002333.
Marek’s disease vaccines: implications in lymphoid
7 Biggs, P.M., and V. Nair. 2012. The long view: 40 years of
organ atrophy and protection. Avian Pathol. 40:573–579.
Marek’s disease research and Avian Pathology. Avian
15 Fadly, A.M., and V. Nair. 2008. Leukosis/Sarcoma
Pathol. 41:3–9.
Group. In: Diseases of Poultry, 12th, Y.M. Saif, et al.,
8 Gimeno, I.M. 2008. Marek’s disease vaccines: a solution
eds. Blackwell Publishing Ltd, Ames. 514–568.
for today but a worry for tomorrow? Vaccine. 26 Suppl 3:
16 Payne, L.N., and V. Nair. 2012. The long view: 40 years
C31–41.
of avian leukosis research. Avian Pathol. 41:1–9.
9 Witter, R.L. 2001. Marek’s disease vaccines—past,
17 Payne, L.N., and K. Venugopal. 2000. Neoplastic
present and future (chicken vs virus—a battle of the
diseases: Marek’s disease, avian leukosis and
centuries). In: Current Progress on Marek’s Disease
reticuloendotheliosis. Rev Sci Tech. 19:544–564.
658 Section II  Viral Diseases

18 Payne, L.N. 1998. Retrovirus‐induced disease in 4 Abdul‐Careem, M.F, B.D Hunter, A.J Sarson, P. Parvizi,
poultry. Poult Sci. 77:1204–1212. H.R Haghighi, L.R Read, M. Heidari, and S. Sharif.
19 Payne, L.N. 1992. Biology of avian retroviruses. In J. 2008. Host responses are induced in feathers of
Levy (ed.). The Retroviridae, Plenum Press: New York. chickens infected with Marek’s disease virus. Virology.
vol 1:299–404. 370:323–332.
20 Payne, L.N. 2000. History of ALV‐J. In E.F Kaleta, L.N 5 Abdul‐Careem, M.F, D.B Hunter, S. Shanmuganathan,
Payne, and U. Heffels‐Redmann (eds.). Proceedings, H.R Haghighi, L. Read, M. Heidari, and S. Sharif. 2008.
International Symposium on ALV‐J and Other Avian Cellular and cytokine responses in feathers of chickens
Retroviruses. Rauischholzhausen, Germany. 3–12. vaccinated against Marek’s disease. Vet Immunol
21 Gao, Y., B. Yun, L. Qin, W. Pan, Y. Qu, Z. Liu, Y. Wang, Immunopathol. 126:362–366.
X. Qi, H. Gao, and X. Wang. 2012. Molecular 6 Abdul‐Careem, M.F, A. Javaheri‐Vayeghan, S.
epidemiology of avian leukosis virus subgroup J in layer Shanmuganathan, H.R Haghighi, L.R Read, K. Haq, D.B
flocks in china. J Clin Microbiol. 50:953–960. Hunter, K.A Schat, M. Heidari, and S. Sharif. 2009.
22 Gao, Y.L., L.T. Qin, W. Pan, Y.Q. Wang, X. Le Qi, H.L. Establishment of an aerosol‐based Marek’s disease
Gao, and X.M. Wang. 2010. Avian leukosis virus virus infection model. Avian Dis.. 53:387–391.
subgroup J in layer chickens, China. Emerg Infect Dis. 7 Abplanalp, H., K.A Schat, and B.W Calnek. 1985.
16:1637–1638. Resistance to Marek’s disease of congenic lines differing
23 Cui, Z., Y. Du, Z. Zhang, and R.F. Silva. 2003. in major histocompatibility haplotypes to 3 virus
Comparison of Chinese field strains of avian leukosis strains. In: Proc. Int. Symp. Marek’s Dis. B.W Calnek
subgroup J viruses with prototype strain HPRS‐103 and and J.L Spencer, eds. American Association of Avian
United States strains. Avian Dis. 47:1321–1330. Pathologists, Kennett Square, PA. 347–358.
24 Nakamura, Y. 2017. Avian biotechnology. Adv Exp Med 8 Abujoub, A., and P.M Coussens. 1995. Development of
Biol. 1001:187–214. a sustainable chick cell line infected with Marek’s
25 Nair, V., and A. Fadly. 2013. Leukosis/Sarcoma group. disease virus. Virology. 214:541–549.
In: Diseases of Poultry, 13th, D.E. Swayne, et al., eds. 9 Abujoub, A.A, D.L Williams, and J.D Reilly. 1999.
John Wiley & Sons, Inc., Ames, Iowa. 553–592. Development of a cell line system susceptible to
26 Gilmore, T.D., and F.S. Wolenski. 2012. NF‐κB: where infection with vaccine strains of MDV. Acta Virol.
did it come from and why? Immunol Rev. 246:14–35. 43:186–191.
27 Zimber, A., K. Perk, M. Ianconescu, Y. Yegana, A. 10 Adldinger, H.K, and B.W Calnek. 1973. Pathogenesis of
Gazit, and A. Yaniv. 1983. Lymphoproliferative disease Marek’s disease: early distribution of virus and viral
of turkeys: pathogenesis, viraemia and serum protein antigens in infected chickens. J Natl Cancer Inst.
analysis following infection. Avian Pathol. 12:101–116. 50:1287–1298.
28 Witter, R.L., I.M. Gimeno, and A.M. Fadly 2005. 11 Afonso, C.L, E.R Tulman, Z. Lu, L. Zsak, D.L Rock, and
Differential diagnosis of lymphoid and myeloid tumors G.F Kutish. 2001. The genome of turkey herpesvirus.
in the chicken, American Association of Avian J Virol. 75:971–978.
Pathologists Athens, GA 27. 12 Aly, M.M, R.L Witter, and A.M Fadly. 1996.
29 Biggs, P.M. 1962. Some observations on the properties Enhancement of reticuloendotheliosis virus‐induced
of cells from lesions of Marek’s disease and lymphoid bursal lymphomas by serotype 2 Marek’s disease virus.
leucosis. In: 13th Symposium of Colston Research Society. Avian Path. 25:81–94.
13 Ameli, H., J.S Gavora, J.L Spencer, and R.W Fairfull.
1992. Genetic resistance to two Marek’s disease viruses
Marek’s Disease
and its relationship to production traits in chickens.
1 2013. Proceedings of the Ninth International Symposium Can J Anim Sci. 72:213–225.
on Marek’s disease and Avian Herpesviruses. June 24–28, 14 Amor, S., S. Strassheim, G. Dambrine, S. Remy, D.
2012, Berlin, Germany. Avian Dis. 57:329–571. Rasschaert, and S. Laurent. 2011. ICP27 protein of
Abdul‐Careem, M.F, B.D Hunter, E. Nagy, L.R Read, B.
2 Marek’s disease virus interacts with SR proteins and
Sanei, J.L Spencer, and S. Sharif. 2006. Development of a inhibits the splicing of cellular telomerase chTERT and
real‐time PCR assay using SYBR Green chemistry for viral vIL8 transcripts. J Gen Virol. 92:1273–1278.
monitoring Marek’s disease virus genome load in feather 15 Anderson, A.S, A. Francesconi, and R.W Morgan. 1992.
tips. J Virol Methods. 133:34–40. Complete nucleotide sequence of the Marek’s disease
Abdul‐Careem, M.F, B.D Hunter, A.J Sarson, A.
3 virus ICP4 gene. Virology. 189:657–667.
Mayameei, H. Zhou, and S. Sharif. 2006. Marek’s disease 16 Anderson, A.S, M.S Parcells, and R.W Morgan. 1998.
virus‐induced transient paralysis is associated with The glycoprotein D (US6) homolog is not essential for
cytokine gene expression in the nervous system. Viral oncogenicity or horizontal transmission of Marek’s
Immunol. 19:167–176. disease virus. J Virol. 72:2548–2553.
Chapter 15  Neoplastic Diseases 659

17 Angamuthu, R., S.S Baskaran, D.R D.R Gopal, J. with virus spread and the pathogenesis of Marek’s
Devarajan, and K. Kathaperumal. 2012. Rapid detection disease. Avian Pathol. 28:287–300.
of the Marek’s disease viral genome in chicken feathers 30 Baigent, S.J, V.K Nair, and H. Le Galludec. 2016.
by loop‐mediated isothermal amplification. J Clin Real‐time PCR for differential quantification of CVI988
Microbiol. 50:961–965. vaccine virus and virulent strains of Marek’s disease
18 Avakian, A.P, R.M Poston, F.K Kong, K.R Van Kampen, virus. J Virol Methods. 233:23–36.
and D.C Tang. 2007. Automated mass immunization of 31 Baigent, S.J, L.J Petherbridge, K. Howes, L.P Smith, R.J
poultry: the prospect for nonreplicating human Currie, and V.K Nair. 2005. Absolute quantitation of
adenovirus‐vectored in ovo vaccines. Expert Rev Marek’s disease virus genome copy number in chicken
Vaccines. 6:457–465. feather and lymphocyte samples using real‐time PCR.
19 Baaten, B.J, K.A Staines, L.P Smith, H. Skinner, T.F J Virol Methods. 123:53–64.
Davison, and C. Butter. 2009. Early replication in 32 Baigent, S.J, L.J Petherbridge, L.P Smith, Y. Zhao, P.M
pulmonary B cells after infection with Marek’s disease Chesters, and V.K Nair. 2006. Herpesvirus of turkey
herpesvirus by the respiratory route. Viral Immunol. reconstituted from bacterial artificial chromosome
22:431–444. clones induces protection against Marek’s disease.
20 Bacon, L.D, H.D Hunt, and H.H Cheng. 2001. Genetic J Gen Virol. 87:769–776.
resistance to Marek’s disease. Curr Top Microbiol 33 Baigent, S.J, L.P Smith, R.J Currie, and V.K Nair. 2007.
Immunol. 255:121–141. Correlation of Marek’s disease herpesvirus vaccine
21 Bacon, L.D, and R.L Witter. 1992. Influence of turkey virus genome load in feather tips with protection, using
herpesvirus vaccination on the B‐haplotype effect on an experimental challenge model. Avian Pathol.
Marek’s disease resistance in 15.B‐congenic chickens. 36:467–474.
Avian Dis. 36:378–385. 34 Baigent, S.J, L.P Smith, L.J Petherbridge, and V.K Nair.
22 Bacon, L.D, and R.L Witter. 1994. B haplotype influence 2011. Differential quantification of cloned CVI988
on the relative efficacy of Marek’s disease vaccines in vaccine strain and virulent RB‐1B strain of Marek’s
commercial chickens. Poult Sci. 73:481–487. disease viruses in chicken tissues, using real‐time PCR.
23 Bacon, L.D, and R.L Witter. 1995. Efficacy of Marek’s Res. Vet. Sci. 91:167–174.
disease vaccines in Mhc heterozygous chickens: Mhc 35 Barrow, A., and K. Venugopal. 1999. Molecular
congenic x inbred line F1 matings. J Hered. 86:269–273. characteristics of very virulent European MDV isolates.
24 Bacon, L.D, R.L Witter, and A.M Fadly. 1989. Acta Virol. 43:90–93.
Augmentation of retrovirus‐induced lymphoid leukosis 36 Barrow, A.D, S.C Burgess, S.J Baigent, K. Howes, and
by Marek’s disease herpesviruses in white leghorn V.K Nair. 2003. Infection of macrophages by a
chickens. J Virol. 63:504–512. lymphotropic herpesvirus: a new tropism for Marek’s
25 Bacon, L.D, R.L Witter, and R.F Silva. 2001. disease virus. J Gen Virol. 84:2635–2645.
Characterization and experimental reproduction of 37 Barrow, A.D, S.C Burgess, K. Howes, and V.K Nair.
peripheral neuropathy in white leghorn chickens. Avian 2003. Monocytosis is associated with the onset of
Pathol. 30:487–499. leukocyte and viral infiltration of the brain in chickens
26 Bader, S.R, S. Kothlow, S. Trapp, S.C Schwarz, H.C infected with the very virulent Marek’s disease virus
Philipp, S. Weigend, A.R Sharifi, R. Preisinger, W. strain C12/130. Avian Pathol. 32:183–191.
Schmahl, B. Kaspers, and K. Matiasek. 2010. Acute 38 Barrow, A.D, S.C Burgess, K. Howes, and K. Venugopal.
paretic syndrome in juvenile White Leghorn chickens 2001. Invasion of avian macrophages by highly virulent
resembles late stages of acute inflammatory Marek’s disease virus strain C12/130 represents a
demyelinating polyneuropathies in humans. “tropic” shift in the pathogenesis. In: Current Progress
J Neuroinflammation. 7:7. on Marek’s Disease Research. K.A Schat, R.M Morgan,
27 Baigent, S., and F. Davison. 2004. Marek’s disease virus: M.S Parcells and J.L Spencer, eds. American
biology and life cycle. In: Marek’s Disease, an Evolving Association of Avian Pathologists, Kennett Square, PA.
Problem. F. Davison and V. Nair, eds. Academic Press, 63–67.
London. 62–77. 39 Baxendale, W. 1969. Preliminary observations on
28 Baigent, S., M. Jamli, A.V Turner, S.C Gilbert, and V. Marek’s disease in ducks and other avian species. Vet
Nair. Replication Defective Adenovirus Serotype 5 Rec. 85:341–342.
Expressing Marek’s Disease Virus Envelope 2016. 40 Beasley, J.N, and J.L Lancaster. 1971. Studies on the role
Glycoprotein as a Potential Marek’s Disease Vaccine in of arthropods as vectors of Marek’s disease. Poult Sci.
Chicken. J Vet Med Res. 3:1049. 50:1552–1552.
29 Baigent, S.J, and T.F Davison. 1999. Development and 41 Beasley, J.N, L.T Patterson, and D.H McWade. 1970.
composition of lymphoid lesions in the spleens of Transmission of Marek’s disease by poultry house dust
Marek’s disease virus‐infected chickens: association and chicken dander. Am J Vet Res. 31:339–344.
660 Section II  Viral Diseases

42 Becker, Y., Y. Asher, E. Tabor, I. Davidson, M. virus BamHI‐H gene family: Genes of putative
Malkinson, and Y. Weisman. 1992. Polymerase chain importance for tumor induction. J Virol.
reaction for differentiation between pathogenic and 63:2534–2542.
non‐pathogenic serotype 1 Marek’s disease viruses 59 Brewer, R.N, W.M Reid, H. Botero, and S.C Schmittle.
(MDV) and vaccine viruses of MDV‐serotypes 2 and 1968. Studies on acute Marek’s disease. 2. The role of
3. J Virol Methods. 40:307–322. coccidia in transmission and induction. Poult Sci.
43 Benton, W.J, and M.S Cover. 1957. The increased 47:2003–2012.
incidence of visceral lymphomatosis in broiler and 60 Brewer, R.N, W.M Reid, J. Johnson, and S.C Schmittle.
replacement birds. Avian Dis. 1:320–327. 1969. Studies on the acute Marek’s disease. VIII. The
44 Biggs, P.M 1966. Avian leukosis and Marek’s disease. role of mosquitoes in transmission under
In: Thirteenth World’s Poultry Congress Symposium experimental conditions. Avian Dis. 13:83–88.
Papers. 91–118. 61 Briles, W.E, R.W Briles, R.E Taffs, and H.A Stone.
45 Biggs, P.M 1968. Marek’s disease. Current state of 1983. Resistance to a malignant lymphoma in chickens
knowledge. Curr Top Microbiol Immunol. 43:93–125. is mapped to subregion of major histocompatibility
46 Biggs, P.M 1985. Spread of Marek’s disease. In: (B) complex. Science. 219:977–979.
Marek’s Disease. L.N Payne, ed. Martinus Nijhoff, 62 Brown, A.C, S.J Baigent, L.P Smith, J.P Chattoo, L.J
Boston, MA. pp 329–340. Petherbridge, P. Hawes, M.J Allday, and V. Nair. 2006.
47 Biggs, P.M 2001. The history and biology of Marek’s Interaction of MEQ protein and C‐terminal‐binding
disease virus. Curr Top Microbiol Immunol. 255:1–24. protein is critical for induction of lymphomas by
48 Biggs, P.M 2004. Marek’s disease—long and difficult Marek’s disease virus. Proc Natl Acad Sci USA.
beginnings. In: Marek’s Disease. An Evolving Problem. 103:1687–1692.
F. Davison and V. Nair, eds. Elsevier Academic Press, 63 Brown, A.C, V. Nair, and M.J Allday. 2012. Epigenetic
London. pp 8–16. regulation of the latency‐associated region of Marek’s
49 Biggs, P.M, and B.S Milne. 1971. Use of the disease virus (MDV) in tumour‐derived T‐cell lines
embryonating egg in studies on Marek’s disease. Am J and primary lymphoma. J Virol.. 86:1683–1695.
Vet Res. 32:1795–1809. 64 Brown, A.C, L.P Smith, L. Kgosana, S.J Baigent, V.
50 Biggs, P.M, and B.S Milne. 1972. Biological properties Nair, and M.J Allday. 2009. Homodimerization of the
of a number of Marek’s disease virus isolates. In: Meq viral oncoprotein is necessary for induction of
Oncogenesis and Herpesviruses. P.M Biggs, G. de The T‐cell lymphoma by Marek’s disease virus. J Virol.
and L.N Payne, eds. IARC, Lyon, France. pp 88–94. 83:11142–11151.
51 Biggs, P.M, and V. Nair. 2012. The long view: 40 years 65 Bruckdorfer, R. 2005. The basics about nitric oxide.
of Marek’s disease research and Avian Pathology. Mol Aspects Med. 26:3–31.
Avian Pathol. 41:3–9. 66 Bublot, M., A. Hurley‐Bacon, T. Hughes, M. Sae Silva,
52 Biggs, P.M, and L.N Payne. 1967. Studies on Marek’s T. Mebatsion, P. Linz, and N. Pritchard. 2016.
disease. I. Experimental transmission. J Natl Cancer Development of a new safe and efficacious Marek’s
Inst. 39:267–280. disease vaccine containing a REV LTR insert in its
53 Biggs, P.M, R.F.W. Shilleto, A.M Lawn, and D.M genome. In: 11th International Symposium on Marek’s
Cooper. 1982. Idiopathic polyneuritis in SPF chickens. disease and avian herpesviruses. C. Desnesvre, M.
Avian Dis. 11:163–178. Bublot and J.F Vautherot, eds., Tours, France. 51.
54 Blake‐Dyke, C., and S. Baigent. 2013. Marek’s disease 67 Bublot, M., and J. Sharma. 2004. Vaccination against
in commercial turkey flocks. Vet Rec. 173. Marek’s disease. In: Marek’s Disease: An Evolving
55 Blankert, J.J, G.A Albers, W.E Briles, M. Vrielink‐van Problem. F. Davison and V. Nair, eds. Elsevier
Ginkel, A.J Groot, G.P. te Winkel, M.G Tilanus, and Academic Press, London. 168–185.
A.J van der Zijpp. 1990. The effect of serologically 68 Buckmaster, A.E, S.D Scott, M.J Sanderson, M.E G.
defined major histocompatibility complex haplotypes Boursnell, L.J N. Ross, and M.M Binns. 1988. Gene
on Marek’s disease resistance in commercially bred sequence and mapping data from Marek’s disease
White Leghorn chickens. Avian Dis. 34:818–823. virus and herpesvirus of turkeys implications for
56 Boodhoo, N., A. Gurung, S. Sharif, and S. Behboudi. herpesvirus classification. J Gen Virol.
2016. Marek’s disease in chickens: a review with focus 69:2033–2042.
on immunology. Vet Res. 47:119. 69 Bumstead, J.M, and L.N Payne. 1987. Production of an
57 Boots, M. 2015. The need for evolutionarily rational immune suppressor factor by Marek’s disease
disease interventions: vaccination can select for lymphoblastoid cell lines. Vet Immunol
higher virulence. PLoS Biol. 13:e1002236. Immunopathol. 16:47–66.
58 Bradley, G., M. Hayashi, G. Lancz, A. Tanaka, and M. 70 Bumstead, N. 1998. Genomic mapping of resistance to
Nonoyama. 1989. Structure of the Marek’s disease Marek’s disease. Avian Pathol. 27:S78–S81.
Chapter 15  Neoplastic Diseases 661

71 Bumstead, N., and J. Kaufman. 2004. Genetic resistance determining the clinical outcome of infection with a
to Marek’s disease. In: Marek’s Disease: An Evolving very virulent oncogenic herpesvirus, Marek’s disease
Problem. F. Davison and V. Nair, eds. Elsevier Academic virus. Avian Pathol. 36:93–99.
Press, London. 112–125. 85 Buza, J.J, and S.C Burgess. 2007. Modeling the
72 Bumstead, N., J. Sillibourne, M. Rennie, N. Ross, and F. proteome of a Marek’s disease transformed cell line: a
Davison. 1997. Quantification of Marek’s disease virus natural animal model for CD30 overexpressing
in chicken lymphocytes using the polymerase chain lymphomas. Proteomics. 7:1316–1326.
reaction with fluorescence detection. J Virol Methods. 86 Calnek, B.W 1972. Effects of passive antibody on early
65:75–81. pathogenesis of Marek’s disease. Infect Immun. 6:193–198.
73 Burgess, S.C 2004. Marek’s disease lymphomas. In: 87 Calnek, B.W 1979. Personal communication.
Marek’s Disease: An Evolving Problem. F. Davison and 88 Calnek, B.W 1985. Pathogenesis of Marek’s disease: a
V. Nair, eds. Elsevier Academic Press, London. 98–111. review. In: Proc Int Symp Marek’s Dis. B.W Calnek and
74 Burgess, S.C 2004. Proteomics in the chicken: tools for J.L Spencer, eds. American Association of Avian
understanding immune responses to avian diseases. Pathologists, Kennett Square, PA. 374–390.
Poult Sci. 83:552–573. 89 Calnek, B.W 1986. Marek’s disease–a model for
75 Burgess, S.C, B.H Basaran, and T.F Davison. 2001. herpesvirus oncology. CRC Crit Rev Microbiol.
Resistance to Marek’s disease herpesvirus‐induced 12:293–320.
lymphoma is multiphasic and dependent on host 90 Calnek, B.W 2001. Pathogenesis of Marek’s disease
genotype. Vet Pathol. 38:129–142. virus infection. Curr Top Microbiol Immunol.
76 Burgess, S.C, and T.F Davison. 1999. A quantitative 255:25–55.
duplex PCR technique for measuring amounts of cell‐ 91 Calnek, B.W, and H.K Adldinger. 1971. Some
associated Marek’s disease virus: differences in two characteristics of cell‐free preparations of Marek’s
populations of lymphoma cells. J Virol Methods. disease virus. Avian Dis. 15:508–517.
82:27–37. 92 Calnek, B.W, H.K Adldinger, and D.E Kahn. 1970.
77 Burgess, S.C, and T.F Davison. 2002. Identification of Feather follicle epithelium: a source of enveloped and
the neoplastically transformed cells in Marek’s disease infectious cell‐free herpesvirus from Marek’s disease.
herpesvirus‐induced lymphomas: recognition by the Avian Dis. 14:219–233.
monoclonal antibody AV37. J Virol. 76:7276–7292. 93 Calnek, B.W, J.C Carlisle, J. Fabricant, K.K Murthy, and
78 Burgess, S.C, J.R Young, B.J G. Baaten, L. Hunt, L.N J. K.A Schat. 1979. Comparative pathogenesis studies
Ross, M.S Parcells, P.M Kumar, C.A Tregaskes, L.F Lee, with oncogenic and nononcogenic Marek’s disease
and T.F Davison. 2004. Marek’s disease is a natural viruses and turkey herpesvirus. Am J Vet Res.
model for lymphomas overexpressing Hodgkin’s disease 40:541–548.
antigen (CD30). Proc Natl Acad Sci USA. 94 Calnek, B.W, J. Fabricant, K.A Schat, and K.K Murthy.
101:13879–13884. 1977. Pathogenicity of low‐virulence Marek’s disease
79 Burgoyne, G.H, and R.L Witter. 1973. Effect of passively viruses in normal versus immunologically
transferred immunoglobulins on Marek’s disease. Avian compromised chickens. Avian Dis. 21:346–358.
Dis. 17:824–837. 95 Calnek, B.W, R.W Harris, C. Buscaglia, K.A Schat, and
80 Burnside, J., and R.W Morgan. Genomics and 2007. B. Lucio. 1998. Relationship between the
Marek’s disease virus. Cytogenet Genome Res. immunosuppressive potential and the pathotype of
117:376–387. Marek’s disease virus isolates. Avian Dis. 42:124–132.
81 Buscaglia, C., and B.W Calnek. 1988. Maintenance of 96 Calnek, B.W, and S.B Hitchner. 1969. Localization of
Marek’s disease herpesvirus latency in vitro by a factor viral antigen in chickens infected with Marek’s disease
found in conditioned medium. J Gen Virol. herpesvirus. J Natl Cancer Inst. 43:935–949.
69:2809–2818. 97 Calnek, B.W, and S.B Hitchner. 1973. Survival and
82 Buscaglia, C., B.W Calnek, and K.A Schat. 1988. disinfection of Marek’s disease virus and the
Effect of immunocompetence on the establishment and effectiveness of filters in preventing airborne
maintenance of latency with Marek’s disease dissemination. Poult Sci. 52:35–43.
herpesvirus. J Gen Virol. 69:1067–1077. 98 Calnek, B.W, S.B Hitchner, and H.K Adldinger. 1970.
83 Buscaglia, C., P. Nervi, and M. Risso. 2004. Lyophilization of cell‐free Marek’s disease herpesvirus
Characterization of four very virulent Argentinian and a herpesvirus from turkeys. Appl Microbiol.
strains of Marek’s disease virus and the influence of one 20:723–726.
of those isolates on synergism between Marek’s disease 99 Calnek, B.W, B. Lucio, K.A Schat, and H.S Lillehoj.
virus. Avian Pathol. 33:190–195. 1989. Pathogenesis of Marek’s disease virus‐induced
84 Butter, C., K. Staines, B. Baaten, L. Smith, and T.F local lesions. 1. Lesion characterization and cell line
Davison. 2007. Route of challenge is critical in establishment. Avian Dis. 33:291–302.
662 Section II  Viral Diseases

100 Calnek, B.W, K.K Murthy, and K.A Schat. 1978. 113 Carvallo, F.R, R.A French, K. Gilbert‐Marcheterre, G.
Establishment of Marek’s disease lymphoblastoid cell Risatti, J.R Dunn, F. Forster, M. Kiupel, and J.A Smyth.
lines from transplantable versus primary lymphomas. 2011. Mortality of one‐week‐old chickens during
Int J Cancer. 21:100–107. naturally occurring Marek’s disease virus infection.
101 Calnek, B.W, K.A Schat, and J. Fabricant. 1980. Vet Pathol. 48:993–998.
Modification of Marek’s disease pathogenesis by in 114 Cebrian, J., C. Kaschka‐Dierich, N. Berthelot, and P.
ovo infection or prior vaccination. In: Viruses in Sheldrick. 1982. Inverted repeat nucleotide sequences
Naturally Occurring Cancers. M. Essex, G. Todaro and in the genomes of Marek’s disease virus and the
H. zur Hausen, eds. Cold Spring Harbor, New York. herpesvirus of the turkey. Proc Natl Acad Sci USA.
185–197. 79:555–558.
102 Calnek, B.W, K.A Schat, E.D Heller, and C. Buscaglia. 115 Chang, S., Z. Ding, J.R Dunn, L.F Lee, M. Heidari, J.
1985. In vitro infection of T‐lymphoblasts with Marek’s Song, C.W Ernst, and H. Zhang. 2011. A comparative
disease virus. In: Proc Int Symp Marek’s Dis. B.W evaluation of the protective efficacy of rMd5deltaMeq
Calnek and J.L Spencer, eds. American Association of and CVI988/ Rispens against a vv+ strain of Marek’s
Avian Pathologists, Kennett Square, PA. 173–187. disease virus infection in a series of recombinant
103 Calnek, B.W, K.A Schat, M.C Peckham, and J. congenic strains of White Leghorn chickens. Avian
Fabricant. 1983. Field trials with a bivalent vaccine Dis. 55:384–390.
(HVT and SB‐1) against Marek’s disease. Avian Dis. 116 Chang, S., J.R Dunn, M. Heidari, L.F Lee, J. Song, C.W
27:844–849. Ernst, Z. Ding, L.D Bacon, and H. Zhang. 2010.
104 Calnek, B.W, K.A Schat, L.J Ross, and C.L Chen. 1984. Genetics and vaccine efficacy: host genetic variation
Further characterization of Marek’s disease virus‐ affecting Marek’s disease vaccine efficacy in White
infected lymphocytes. II. In vitro infection. Int J Leghorn chickens. Poult Sci. 89:2083–2091.
Cancer. 33:399–406. 117 Chang, S., Q. Xie, J.R Dunn, C.W Ernst, J. Song, and
105 Calnek, B.W, K.A Schat, L.J Ross, W.R Shek, and C.L H. Zhang. 2014. Host genetic resistance to Marek’s
Chen. 1984. Further characterization of Marek’s disease sustains protective efficacy of herpesvirus of
disease virus‐infected lymphocytes. I. In vivo turkey in both experimental and commercial lines of
infection. Int J Cancer. 33:389–398. chickens. Vaccine. 32:1820–1827.
106 Calnek, B.W, K.A Schat, W.R Shek, and C.‐L.H Chen. 118 Chattoo, J.P, M.P Stevens, and V. Nair. 2006. Rapid
1982. In vitro infection of lymphocytes with Marek’s identification of non‐essential genes for in vitro
disease virus. J Natl Cancer Inst. 69:709–713. replication of Marek’s disease virus by random
107 Calnek, B.W, W.R Shek, and K.A Schat. 1981. Latent transposon mutagenesis. J Virol Methods.
infections with Marek’s disease virus and turkey 135:288–291.
herpesvirus. J Natl Cancer Inst. 66:585–590. 119 Chbab, N., A. Egerer, I. Veiga, K.W Jarosinski, and N.
108 Calnek, B.W, W.R Shek, and K.A Schat. 1981. Osterrieder. 2010. Viral control of vTR expression is
Spontaneous and induced herpesvirus genome critical for efficient formation and dissemination of
expression in Marek’s disease tumor cell lines. Infect lymphoma induced by Marek’s disease virus (MDV).
Immun. 34:483–491. Vet Res. 41:56.
109 Calnek, B.W, W.R Shek, K.A Schat, and J. Fabricant. 120 Cheng, H.H, P. Kaiser, and S.J Lamont. 2013. Integrated
1982. Dose‐dependent inhibition of virus rescue from genomic approaches to enhance genetic resistance in
lymphocytes latently infected with turkey herpesvirus chickens. Ann Rev Anim Biosci. 1:239–260.
or Marek’s disease virus. Avian Dis. 26:321–331. 121 Cheng, H.H, S. Perumbakkam, A.B Pyrkosz, J.R Dunn,
110 Calnek, B.W, T. Ubertini, and H.K Adldinger. 1970. A. Legarra, and W.M Muir. 2015. Fine mapping of
Viral antigen, virus particles, and infectivity of tissues QTL and genomic prediction using allele‐specific
from chickens with Marek’s disease. J Natl Cancer expression SNPs demonstrates that the complex trait
Inst. 45:341–351. of genetic resistance to Marek’s disease is
111 Calnek, W., and M.W Smith. 1972. Vaccination predominantly determined by transcriptional
against Marek’s disease with cell‐free turkey regulation. BMC Genomics. 16:816.
herpesvirus: interference by maternal antibody. Avian 122 Cho, B.R 1975. Horizontal transmission of turkey
Dis. 16:954–957. herpesvirus to chickens. IV. Viral maturation in the
112 Cao, W., J. Mays, G. Kulkarni, J. Dunn, R.M Fulton, feather follicle epithelium. Avian Dis. 19:136–141.
and A. Fadly. 2015. Further observations on serotype 123 Cho, B.R 1976. A possible association between plaque
2 Marek’s disease virus‐induced enhancement of type and pathogenicity of Marek’s disease herpesvirus.
spontaneous avian leukosis virus‐like bursal Avian Dis. 20:324–331.
lymphomas in ALVA6 transgenic chickens. Avian 124 Cho, B.R 1977. Dual virus maturation of both
Pathol. 44:23–27. pathogenic and apathogenic Marek’s disease
Chapter 15  Neoplastic Diseases 663

herpesvirus (MDHV) in the feather follicles of dually 139 Coussens, P.M, and L.F Velicer. 1988. Structure and
infected chickens. Avian Dis. 21:501–507. complete nucleotide sequence of the Marek’s disease
125 Cho, B.R, and S.G Kenzy. 1972. Isolation and herpesvirus gp57–65 gene. J Virol. 62:2373–2379.
characterization of an isolate (HN) of Marek’s disease 140 Couteaudier, M., K. Courvoisier, L. Trapp‐Fragnet, C.
virus with low pathogenicity. Appl Microbiol. Denesvre, and J. Vautherot. 2016. Keratinocytes
24:299–306. derived from chicken embryonic stem cells support
126 Cho, K.‐O., M. Mubarak, T. Kimura, K. Ochiai, and C. Marek’s disease virus infection: a highly differentiated
Itakura. 1996. Sequential skin lesions in chickens cell model to study viral replication and
experimentally infected with Marek’s disease virus. morphogenesis. Virol J. 13.
Avian Pathol. 25:325–343. 141 Couteaudier, M., K. Courvoisier, L. Trapp‐Fragnet, C.
127 Cho, K.‐O., K. Ochiai, Y. Fukikawa, and C. Itakura. Denesvre, and J.F Vautherot. 2016. Keratinocytes
1997. Cutaneous lesions in broiler chickens derived from chicken embryonic stem cells support
spontaneously afftected with Marek’s disease. Avian Marek’s disease virus infection: a highly differentiated
Pathol. 26:277–291. cell model to study viral replication and
128 Cho, K.O, N.Y Park, D. Endoh, K. Ohashi, C. morphogenesis. Virol J. 13:7.
Sugimoto, C. Itakura, and M. Onuma. 1998. Cytology 142 Couteaudier, M., and C. Denesvre. 2014. Marek’s
of feather pulp lesions from Marek’s disease (MD) disease virus and skin interactions. Vet Res. 45:36.
virus‐infected chickens and its application for 143 Crittenden, L.B, R.L Muhm, and B.R Burmester. 1972.
diagnosis and prediction of MD. J Vet Med Sci. Genetic control of susceptibility to the avian leukosis
60:843–847. complex. 2. Marek’s disease. Poult Sci. 51:261–267.
129 Chubb, R.C, and A.E Churchill. 1969. Effect of 144 Cui, X., L.F Lee, H.D Hunt, W.M Reed, B. Lupiani, and
maternal antibody on Marek’s disease. Vet Rec. S.M Reddy. A 2005. Marek’s disease virus vIL‐8
85:303–305. deletion mutant has attenuated virulence and confers
130 Churchill, A.E 1985. Production of vaccines. In: protection against challenge with a very virulent plus
Marek’s Disease, Scientific Basis and Methods of strain. Avian Dis. 49:199–206.
Control. L.N Payne, ed. Martinus Nijhoff Boston, MD. 145 Cui, X., L.F Lee, W.M Reed, H.J Kung, and S.M Reddy.
251–266. 2004. Marek’s disease virus‐encoded vIL‐8 gene is
131 Churchill, A.E, R.C Chubb, and W. Baxendale. 1969. involved in early cytolytic iInfection but dispensable
The attenuation, with loss of oncogenicity of the for establishment of latency. J Virol. 78:4753–4760.
herpes‐type virus of Marek’s disease (strain HPRS‐16) 146 Cui, Z.‐Z., Y. Ding, and L.F Lee. 1990. Marek’s disease
on passage in cell culture. J Gen Virol. 4:557–564. virus gene clones encoding virus‐specific
132 Churchill, A.E, L.N Payne, and R.C Chubb. 1969. phosphorylated polypeptides and serological
Immunization against Marek’s disease using a live characterization of fusion proteins. Virus Genes.
attenuated virus. Nature. 221:744–747. 3:309–322.
133 Cole, R.K 1968. Studies on genetic resistance to 147 Cui, Z., A. Qin, X. Cui, Y. Du, and L.F Lee. 2001.
Marek’s disease. Avian Dis. 12:9–28. Molecular identification of 3 epitopes on 38kd
134 Colmano, G., and W.B Gross. 1971. Effect of phosporylated proteins of Marek’s disease viruses. In:
metyrapone and DDD on infectious diseases. Poult Current Progress on Marek’s Disease Research. K.A
Sci. 50:850–854. Schat, R.W Morgan, M.S Parcells and J.L Spencer, eds.
135 Colwell, W.M, C.F Simpson, L.E Williams, Jr., and D.J American Association of Avian Pathologists, Kennett
Forrester. 1973. Isolation of a herpesvirus from wild Square, PA. 103–108.
turkeys in Florida. Avian Dis. 17:1–11. 148 Cui, Z., A. Qin, L.F Lee, P. Wu, and H.J Kung. 1999.
136 Cortes, A.L, E.R Montiel, S. Lemiere, and I.M Construction and characterization of a H19 epitope
Gimeno. 2011. Comparison of blood and feather pulp point mutant of MDV CVI988/Rispens strain. Acta
samples for the diagnosis of Marek’s disease and for Virol. 43:169–173.
monitoring Marek’s disease vaccination by real 149 Cui, Z., G. Zhuang, X. Xu, A. Sun, and S. Su. 2010.
time‐PCR. Avian Dis. 55:302–310. Molecular and biological characterization of a Marek’s
137 Coudert, F., A. Vuillaume, M. Wyers, and A.M disease virus field strain with reticuloendotheliosis
Chaussé. 1997. Marek’s disease in turkeys. World virus LTR insert. Virus Genes. 40:236–243.
Poult. S28–29. 150 Cushing, T.L, K.A Schat, S.L States, J.L Grodio, P.H
138 Coupeau, D., G. Dambrine, and D. Rasschaert. 2012. O’Connell, and E.L Buckles. 2011. Characterization of
The kinetic expression analysis of the cluster mdv1‐ the host response in systemic isosporosis
mir‐M9~M4, genes meq and vIL‐8 differs between (atoxoplasmosis) in a colony of captive American
the lytic and latent phases of Marek’s disease virus goldfinches (Spinus tristis) and house sparrows
infection. J Gen Virol. PMID: 22442112. (Passer domesticus). Vet Pathol. 48:985–992.
664 Section II  Viral Diseases

151 Dandapat, S., H.K Pradhan, and G.C Mohanty. 1994. 165 Ding, J., Z. Cui, S. Jiang, and Y. Li. 2008. Study on the
Anti‐idiotype antibodies to Marek’s disease‐associated structure of heteropolymer pp38/pp24 and its
tumour surface antigen in protection against Marek’s enhancement on the bi‐directional promoter
disease. Vet Immunol Immunopathol. 40:353–366. upstream of pp38 gene in Marek’s disease virus. Sci
152 Dang, L., M. Teng, H.W Li, H.Z Li, S.M Ma, P. Zhao, China Series C Life Sci. 51:821–826.
X.J Li, R.G Deng, G.P Zhang, and J. Luo. Dynamic 166 Djeraba, A., N. Bernardet, G. Dambrine, and P. Quere.
2017. Changes in the Splenic Transcriptome of 2000. Nitric oxide inhibits Marek’s disease virus
Chickens during the Early Infection and Progress of replication but is not the single decisive factor in
Marek’s Disease. Sci Rep. 7:11648. interferon‐gamma‐mediated viral inhibition. Virology.
153 Davidson, I., and R. Borenstein. 1999. Multiple 277:58–65.
infection of chickens and turkeys with avian 167 Djeraba, A., E. Musset, N. Bernardet, Y. Le Vern, and
oncogenic viruses: prevalence and molecular analysis. P. Quere. 2002. Similar pattern of iNOS expression,
Acta Virol. 43:136–142. NO production and cytokine response in genetic and
154 Davidson, I., T. Maray, M. Malkinson, and Y. Becker. vaccination‐acquired resistance to Marek’s disease.
1986. Detection of Marek’s disease virus antigens and Vet Immunol Immunopathol. 85:63–75.
DNA in feathers from infected chickens. J Virol Meth. 168 Djeraba, A., E. Musset, J.W Lowenthal, D.B Boyle,
13:231–244. A.M Chausse, M. Peloille, and P. Quere. 2002.
155 Davidson, I., Y. Weisman, S. Perl, and M. Malkinson. Protective effect of avian myelomonocytic growth
1998. Differential diagnosis of two paralytic factor in infection with Marek’s disease virus. J Virol.
conditions affecting young birds with emphasis on 76:1062–1070.
PCR findings. Avian Pathol. 27:417–419. 169 Doak, R.L, J.F Munnell, and W.L Ragland. 1973.
156 Davison, F., S. Baigent, M. Rennie, and N. Bumstead. Ultrastructure of tumor cells in Marek’s disease
1998. Age‐and strain‐related differences in the virus‐ infected chickens. Am J Vet Res. 34:1063–1069.
quantity of Marek’s disease virus in different sub‐ 170 Dudnikova, E., S. Norkina, A. Vlasov, A. Slobodchuk,
populations of lymphocytes. Avian Pathol. 27:S88. L.F Lee, and R.L Witter. 2007. Evaluation of Marek’s
157 Davison, F., and P. Kaiser. 2004. Immunity to Marek’s disease field isolates by the “best fit” pathotyping
disease. In: Marek’s Disease: An Evolving Problem. F. assay. Avian Pathol. 36:135–143.
Davison and V. Nair, eds. Elsevier Academic Press, 171 Duncan, A.J, and S.J R. Heales. 2005. Nitric oxide and
London. pp 126–141. neurological disorders. Mol Aspects Med. 26:67–96.
158 Davison, F., and V. Nair, eds. 2004. Marek’s disease: an 172 Dunn, J.R, K. Auten, M. Heidari, and C. Buscaglia.
evolving problem. Elsevier Academic Press, London. 2014. Correlation between Marek’s disease virus
159 Davison, F., and V. Nair. 2005. Use of Marek’s disease pathotype and replication. Avian Dis. 58:287–292.
vaccines: could they be driving the virus to increasing 173 Dunn, J.R, A. Black Pyrkosz, and H.H Cheng. 2016.
virulence? Expert Rev Vaccines. 4:77–88. Pathotyping of current Marek’s disease virus field
160 de Boer, G.F, J. Pol, and H. Oei. 1987. Biological strains and identification of sequence variants to
characteristics of Marek’s disease vaccine CVI‐988 predict virulence. In: 11th International Symposium
clone C. Vet Q. 9:16S–28S. on Marek’s disease and Avian Herpesvirus. M. Bublot,
161 de Boer, G.F, J.M A. Pol, and S.H M. Jeurissen. 1989. C. Denesvre, and J.‐F. Vautherot, eds. INRA, Tours,
Marek’s disease vaccination strategies using vaccines France. 10.
made from three avian herpesvirus serotypes. In: 174 Dunn, J.R, and I.M Gimeno. Current 2013. Status of
Advances in Marek’s Disease Research. S. Kato, T. Marek’s disease in the United States and worldwide
Horiuchi, T. Mikami and K. Hirai, eds. Japanese based on a questionnaire survey. Avian Dis. 57:483–490.
Association on Marek’s Disease, Osaka, Japan. pp 175 Dunn, J.R, and R.F Silva. 2012. Ability of MEQ‐
405–413. deleted MDV vaccine candidates to adversely affect
162 Delecluse, H.‐J., and W. Hammerschmidt. 1993. Status lymphoid organs and chicken weight gain. Avian Dis.
of Marek’s disease virus in established lymphoma cell 56:494–500.
lines: Herpesvirus integration is common. J Virol. 67:82–92. 176 Dunn, J.R, R.L Witter, R.F Silva, L.F Lee, J. Finlay, B.A
163 Denesvre, C. 2013. Marek’s disease virus Marker, J.B Kaneene, R.M Fulton, and S.D Fitzgerald.
morphogenesis. Avian Dis. 57:340–350. 2010. The effect of the time interval between
164 Denesvre, C., S. Remy, L. Trapp‐Fragnet, L.P Smith, S. exposures on the susceptibility of chickens to
Georgeault, J.F Vautherot, and V. Nair. 2016. Marek’s superinfection with Marek’s disease virus. Avian Dis.
disease virus undergoes complete morphogenesis 54:1038–1049.
after reactivation in a T‐lymphoblastoid cell line 177 Eidson, C.S, O.J Fletcher, and D.P Anderson. 1971.
transformed by recombinant fluorescent marker virus. Characterization of Ala‐1 isolate of acute Marek’s
J Gen Virol. 97:480–486. disease. Poult Sci. 50:693–699.
Chapter 15  Neoplastic Diseases 665

178 Eidson, C.S, R.K Page, and S.H Kleven. 1978. 191 Fabricant, C.G. 1985. Atherosclerosis: The
Effectiveness of cell‐free or cell‐associated turkey consequence of infection with a herpesvirus. Adv Vet
herpesvirus vaccine against Marek’s disease in Sci Comp Med. 30:39–66.
chickens as influenced by maternal antibody, vaccine 192 Fabricant, C.G, J. Fabricant, M.M Litrenta, and C.R
dose, and time of exposure to Marek’s disease virus. Minick. 1978. Virus‐induced atherosclerosis. J Exp
Avian Dis. 22:583–597. Med. 148:335–340.
179 Eidson, C.S, S.C Schmittle, R.B Goode, and J.B Lai. 193 Fabricant, C.G, D.P Hajjar, C.R Minick, and J.
1966. Induction of leukosis tumors with the beetle Fabricant. 1981. Herpesvirus infection enhances
Alphitobius diaperinus. Am J Vet Res. 27:1053–1057. cholesterol and cholesteryl ester accumulation in
180 Ekperigin, H.E, A.M Fadly, L.F Lee, X. Liu, and R.H cultured arterial smooth muscle cells. Am J Pathol.
McCapes. 1983. Comb lesions and mortality patterns 105:176–184.
in white leghorn layers affected by Marek’s disease. 194 Fabricant, J., B.W Calnek, and K.A Schat. 1982. The
Avian Dis. 27:503–512. early pathogenesis of turkey herpesvirus infection in
181 Ellis, M.N, C.S Eidson, J. Brown, O.J Fletcher, and S.H chickens and turkeys. Avian Dis. 26:257–264.
Kleven. 1981. Serological responses to mycoplasma 195 Faiz, N., A.L Cortes, J.S Guy, O.J Fletcher, M. West, E.
synoviae in chickens infected with virulent or Montiel, and I.M Gimeno. 2016. Early infection with
avirulent strains of Marek’s disease virus. Poult Sci. Marek’s disease virus can jeopardize protection
60:1344–1347. conferred by laryngotracheitis vaccines: a method to
182 Elmubarak, A.K, J.M Sharma, R.L Witter, and V.L study MDV‐induced immunosuppression. Avian
Sanger. 1982. Marek’s disease in turkeys: Lack of Pathol. 20:1–32.
protection by vaccination. Am J Vet Res. 43:740–742. 196 Faiz, N.M, A.L Cortes, J.S Guy, O.J Fletcher, T.
183 Else, R.W 1974. Vaccinal immunity to Marek’s disease Cimino, and I.M Gimeno. 2017. Evaluation of factors
in bursectomized chickens. Vet Rec. 95:182–187. influencing the development of late Marek’s disease
184 Emara, M.G, M.A Abdellatif, D.L Pollock, M. Sadjadi, virus‐induced immunosuppression: virus pathotype
S.S Cloud, C.R Pope, J.K Rosenberger, and H. Kim. and host sex. Avian Pathol. 1–39.
2001. Genetic variation in susceptibility to Marek’s 197 Faiz, N.M, A.L Cortes, J.S Guy, J.E Fogle, and I.M
disease in a commercial broiler population. Avian Dis. Gimeno. 2016. Efficacy of various Marek’s disease
45:400–409. vaccines protocols for prevention of Marek’s disease
185 Endoh, D. 1996. Enhancement of gene expression by virus‐induced immunosuppression. Vaccine.
Marek’s disease virus homologue of the herpes 34:4180–4187.
simplex virus‐1 ICP4. Jpn J Vet Res. 44:136–137. 198 Ficken, M.D, M.P Nasisse, G.D Boggan, J.S Guy, D.P
186 Engel, A.T, R.K Selvaraj, J.P Kamil, N. Osterrieder, and Wages, R.L Witter, J.K Rosenberger, and R.M
B.B Kaufer. 2012. Marek’s disease viral interleukin‐8 Nordgren. 1991. Marek’s disease virus isolates with
promotes lymphoma formation through targeted unusual tropism and virulence for ocular tissues:
recruitment of B cells and CD4+ CD25+ T cells. J Clinical findings, challenge studies and pathological
Virol. 86:8536–8545. features. Avian Pathol. 20:461–474.
187 Erf, G.F, T.K Bersi, X.L Wang, G.P Sreekumar, and J.R 199 Fletcher, O.J, C.S Eidson, and R.K Page. 1971.
Smyth. 2001. Herpesvirus connection in the Pathogenesis of Marek’s disease induced in chickens
expression of autoimmune vitiligo in Smyth line by contact exposure to GA isolate. Am J Vet Res.
chickens. Pigment Cell Res. 14:40–46. 32:1407–1416.
188 Esaki, M., A. Godoy, J.K Rosenberger, S.C 200 Fletcher, O.J, and L.W Schierman. 1985. Variation in
Rosenberger, Y. Gardin, A. Yasuda, and K.M Dorsey. histology and growth characteristics of transplantable
2013. Protection and antibody response caused by Marek’s disease lymphomas. Cancer Res.
turkey herpesvirus vector Newcastle disease vaccine. 45:1762–1765.
Avian Dis. 57:750–755. 201 Fragnet, L., M.A Blasco, W. Klapper, and D.
189 Esaki, M., L. Noland, T. Eddins, A. Godoy, S. Saeki, S. Rasschaert. 2003. The RNA subunit of telomerase is
Saitoh, A. Yasuda, and K.M Dorsey. 2013. Safety and encoded by Marek’s disease virus. J Virol. 77:5985–5996.
efficacy of a turkey herpesvirus vector 202 Fragnet, L., E. Kut, and D. Rasschaert. 2005.
laryngotracheitis vaccine for chickens. Avian Dis. Comparative functional study of the viral telomerase
57:192–198. RNA based on natural mutations. J Biol Chem.
190 Ewert, D.L Molecular 1997. Approaches for the 280:23502–23515.
diagnosis of avian lymphomas. In: Diagnosis and 203 Friedman, A., E. Shalem‐Meilin, and E.D Heller. 1992.
Control of Neoplastic Diseases of Poultry. A.M Fadly, Marek’s disease vaccines cause temporary B‐
K.A Schat and J.L Spencer, eds. American Association lymphocyte dysfunction and reduced resistance to
of Avian Pathologists, Kennett Square. 12–18. infection in chicks. Avian Pathol. 21:621–631.
666 Section II  Viral Diseases

204 Fukuchi, K., A. Tanaka, L.W Schierman, R.L Witter, against infectious laryngotracheitis and Marek’s
and M. Nonoyama. 1985. The structure of Marek’s disease. Avian Dis. 59:255–262.
disease virus DNA: the presence of unique expansion 216 Gimeno, I.M, A.L Cortes, N.M Faiz, T. Villalobos, H.
in nonpathogenic viral DNA. Proc Natl Acad Sci USA. Badillo, and T. Barbosa. 2016. Efficacy of various HVT
82:751–754. vaccines (conventional and recombinant) against
205 Gardin, Y., V. Palya, K.M Dorsey, J. El‐Attrache, F. Marek’s disease in broiler chickens: effect of dose and
Bonfante, S. Wit, D. Kapczynski, W.H Kilany, F. Rauw, age of vaccination. Avian Dis. 60:662–668.
M. Steensels, and R.D Soejoedono. Experimental and 217 Gimeno, I.M, A.L Cortes, E.R Montiel, S. Lemiere,
Field Results Regarding Immunity 2016. Induced by a and A.K R. Pandiri. 2011. Effect of diluting Marek’s
recombinant turkey herpesvirus H5 vector vaccine disease vaccines on the outcomes of Marek’s disease
against H5N1 and other H5 highly pathogenic avian virus infection when challenged with highly virulent
influenza virus challenges. Avian Dis. 60:232–237. Marek’s disease viruses. Avian Dis. 55:263–272.
206 Geerligs, H., S. Quanz, B. Suurland, I.E Spijkers, J. 218 Gimeno, I.M, A.L Cortes, and R.F Silva. 2008. Load of
Rodenberg, F.G Davelaar, B. Jongsma, and M. Kumar. challenge marek’s disease virus DNA in blood as a
2008. Efficacy and safety of cell associated vaccines criterion for early diagnosis of Marek’s disease
against Marek’s disease virus grown in a continuous tumors. Avian Dis. 52:203–208.
cell line from chickens. Vaccine. 26:5595–5600. 219 Gimeno, I.M, A.L Cortes, R.L Witter, and A.R Pandiri.
207 Geerligs, H.J, and A. Hoogendam. 2007. 2012. Optimization of the protocols for double
Determination of optimal conditions for thawing and vaccination against Marek’s disease using
diluting cell‐bound CVI 988 Marek’s disease vaccine commercially available vaccines: evaluation of
and stability of the diluted vaccine. Avian Dis. protection, vaccine replication, and activation of T
51:969–973. cells. Avian Dis. 56:295–305.
208 Gelb, J., Jr., D.J Jackwood, E.M Brannick, and B.S 220 Gimeno, I.M, J. Dunn, A.L Cortes, A.E El‐Gohari, and
Ladman. 2016. Efficacy of recombinant HVT‐IBD R.F Silva. 2014. Detection and differentiation of
Vaccines administered to broiler chicks from a single CVI988 (Rispens vaccine) from other serotype 1
breeder flock at 30 and 60 weeks of age. Avian Dis. Marek’s disease viruses. Avian Dis. 58 2:232–243.
60:603–612. 221 Gimeno, I.M, N.M Faiz, A.L Cortes, T. Barbosa, T.
209 Gilka, F., and J.L Spencer. 1995. Extravascular Villalobos, and A.R Pandiri. 2015. In ovo vaccination
hemolytic anemia in chicks infected with highly with HVT hasten maturation of chicken embryos
pathogenic Marek’s disease viruses. Avian Pathol. immune responses in specific pathogen free chickens
24:393–410. (SPAFAS). Avian Dis. 59:375–383.
210 Gimeno, I.M 2004. Future strategies for controlling 222 Gimeno, I.M, and A.R Pandiri. 2013. Virus‐induced
Marek’s disease. In: Marek’s Disease. F. Davison and V. immunosuppression: Marek’s disease virus infection
Nair, eds. Elsevier Academic Press, London. 186–199. and associates syndromes. In: Immunosuppresive
211 Gimeno, I.M 2008. Marek’s disease vaccines: a Diseases of Poultry. I.M Gimeno, ed. Servet, Zaragoza.
solution for today but a worry for tomorrow? Vaccine. 124–148.
26:C31–41. 223 Gimeno, I.M, and P.S Wakenell. 2016. Marek’s disease.
212 Gimeno, I.M 2015. Stepwise diagnostic approach to In: A Laboratory Manual for the Isolation,
investigate a Marek’s disease outbreak. In: American Identification and Characterization of Avian
Association of Avian Pathologist Symposium: An Pathogens. S.M Williams, L. Dufour‐Zavala, M.W
update on Marek’s disease vaccination, diagnosis, and Jackwood, M.D Lee, B. Lupiani and W.M Reed, eds.
immunosuppression. A.R Pandiri, ed. American American Association of Avian Pathologists,
Association of Avian Pathologists, Boston, MA. Jacksonville, FL. 249–258.
213 Gimeno, I.M, and A.L Cortes. 2011. Chronological 224 Gimeno, I.M, R.L Witter, A.L Cortes, S.M Reddy, and
study of cytokine transcription in the spleen and lung A.R Pandiri. 2012. Standardization of a model to study
of chickens after vaccination with serotype 1 Marek’s revaccination against Marek’s disease under
disease vaccines. Vaccine. 29:1583–1594. laboratory conditions. Avian Pathol. 41:59–68.
214 Gimeno, I.M, A.L Cortes, N.M Faiz, T. Barbosa, and 225 Gimeno, I.M, R.L Witter, A.L Cortes, and W.M Reed.
T. Villalobos. 2015. Evaluation of factors influencing 2011. Replication ability of three highly protective
efficacy of vaccine strain CVI988 against Marek’s Marek’s disease vaccines: implications in lymphoid
disease in meat‐type chickens. Avian Dis. 59:400–409. organ atrophy and protection. Avian Pathol. 40:573–579.
215 Gimeno, I.M, A.L Cortes, N.M Faiz, B.A Hernandez‐ 226 Gimeno, I.M, R.L Witter, A.M Fadly, and R.F Silva.
Ortiz, J.S Guy, H.D Hunt, and R.F Silva. 2015. 2005. Novel criteria for the diagnosis of Marek’s
Evaluation of the protection efficacy of a serotype disease virus‐induced lymphomas. Avian Pathol.
1 Marek’s disease virus‐vectored bivalent vaccine 34:332–340.
Chapter 15  Neoplastic Diseases 667

227 Gimeno, I.M, R.L Witter, H.D Hunt, L.F Lee, S.M 240 Han, P.F, and J.R Smyth, Jr. 1972. The influence of
Reddy, and U. Neumann. 2001. Marek’s disease virus growth rate on the development of Marek’s disease in
infection in the brain: virus replication, cellular chickens. Poult Sci. 51:975–985.
infiltration, and major histocompatibility complex 241 Handberg, K.J, O.L Nielsen, and P.H Jorgensen. 2001.
antigen expression. Vet Pathol. 38:491–503. The use of serotype 1‐ and serotype 3‐specific
228 Gimeno, I.M, R.L Witter, H.D Hunt, S.M Reddy, L.F polymerase chain reaction for the detection of
Lee, and R.F Silva. 2005. The pp38 gene of Marek’s Marek’s disease virus in chickens. Avian Pathol.
disease virus (MDV) is necessary for cytolytic 30:243–249.
infection of B cells and maintenance of the 242 Haq, K., J.T Brisbin, N. Thanthrige‐Don, M. Heidari,
transformed state but not for cytolytic infection of the and S. Sharif. 2010. Transcriptome and proteome
feather follicle epithelium and horizontal spread of profiling of host responses to Marek’s disease virus
MDV. J Virol. 79:4545–4549. in chickens. Vet Immunol Immunopathol.
229 Gimeno, I.M, R.L Witter, H.D Hunt, S.M Reddy, and 138:292–302.
U. Neumann. 2001. Differential attenuation of the 243 Haq, K., I. Elawadli, P. Parvizi, A.I Mallick, S.
induction by Marek’s disease virus of transient Behboudi, and S. Sharif. 2011. Interferon‐γ influences
paralysis and persistent neurological disease: a model immunity elicited by vaccines against very virulent
for pathogenesis studies. Avian Pathol. 30:397–410. Marek’s disease virus. Antiviral Res. 90:218–226.
230 Gimeno, I.M, R.L Witter, and U. Neumann. 2002. 244 Haq, K., T. Fear, A. Ibraheem, M.F Abdul‐Careem,
Neuropathotyping: a new system to classify Marek’s and S. Sharif. 2012. Influence of vaccination with
disease virus. Avian Dis. 46:909–918. CVI988/Rispens on load and replication of a very
231 Gimeno, I.M, R.L Witter, and W.M Reed. 1999. Four virulent Marek’s disease virus strain in feathers of
distinct neurologic syndromes in Marek’s disease: chickens. Avian Pathol. 41:69–76.
effect of viral strain and pathotype. Avian Dis. 245 Haq, K., K.A Schat, and S. Sharif. 2013. Immunity to
43:721–737. Marek’s disease: where are we now? Dev Comp
232 Godoy, A., A. Icard, M. Martinez, A. Mashchenko, M. Immunol. 41:439–446.
Garcia, and J. El‐Attrachea. 2013. Detection of 246 Haq, K., K.A Schat, and S. Sharif. 2013. Immunity to
infectious laryngotracheitis virus antibodies by Marek’s disease: where are we now? Dev Comp
glycoprotein‐specific ELISAs in chickens vaccinated Immunol. 41:439–446.
with viral vector vaccines. Avian Dis. 57:432–436. 247 Hartmann, W., K. Hala, and G. Heil. The 1992. B
233 Goodchild, W.M 1969. Some observations on Marek’s blood group system of the chicken and resistance to
disease (fowl paralysis). Vet Rec. 84:87–89. Marek’s disease: effect of B blood group genotypes in
234 Groot, A.J C., and G.A A. Albers. 1992. The effect of leghorn crosses. Arch Tierz. 35:169–180.
MHC on resistance to Marek’s disease in White 248 Hearn, C., L. Preeyanon, H.D Hunt, and I.A York.
Leghorn crosses. In: Proc 4th Int Symp Marek’s 2015. An MHC class I immune evasion gene of
disease. G. de Boer and S.H M. Jeurissen, eds. Ponsen Marek’s disease virus. Virology. 475:88–95.
& Looijen, Wageningen, Amsterdam, The 249 Heidari, M., and P.C Delekta. 2017. Transcriptomic
Netherlands. 185–188. analysis of host immune response in the skin of
235 Gross, W.B 1972. Effect of social stress on occurrence chickens infected with Marek’s disease virus. Viral
of Marek’s disease in chickens. Am J Vet Res. Immunol. 30:377–387.
33:2275–2279. 250 Heidari, M., S.D Fitzgerald, H.M Zhang, R.F Silva, L.F
236 Hafner, S., B.G Harmon, G.N Rowland, R.G Stewart, Lee, and J.R Dunn. 2007. Marek’s disease virus‐
and J.R Glisson. 1991. Spontaneous regression of induced skin leukosis in scaleless chickens: tumor
“dermal squamous cell carcinoma” in young chickens. development in the absence of feather follicles. Avian
Avian Dis. 35:321–327. Dis. 51:713–718.
237 Hajjar, D.P, C.G Fabricant, C.R Minick, and J. 251 Heidari, M., A.J Sarson, M. Huebner, S. Sharif, D.
Fabricant. 1986. Virus‐induced atherosclerosis. Kireev, and H. Zhou. 2010. Marek’s disease virus‐
Herpesvirus infection alters aortic cholesterol induced immunosuppression: array analysis of
metabolism and accumulation. Am J Pathol. 122:62–70. chicken immune response gene expression profiling.
238 Halouzka, R., and V. Jurajda. 1992. Pathological Viral Immunol. 23:309–319.
lesions in the organs of chicks after infection with 252 Heidari, M., D. Wang, and S. Sun. 2017. Early immune
turkey herpesvirus THV‐BI0‐I. Vet. Med. (Praha). responses to Marek’s disease vaccines. Viral Immunol.
37:463–470. 30:167–177.
239 Halvorson, D.A, and D.O Mitchel. 1979. Loss of 253 Heidari, M., H.M Zhang, and S. Sharif. 2008. Marek’s
cell‐associated Marek’s disease vaccine titer during disease virus induces Th2 activity during cytolytic
thawing, reconstitution and use. Avian Dis. 23:848–853. infection. Viral Immunol. 21:203–214.
668 Section II  Viral Diseases

254 Heller, E.D, and K.A Schat. 1987. Enhancement of 267 Hunt, H.D, and J.R Dunn. Serial 2011. Transfer of a
natural killer cell activity by Marek’s disease vaccines. transplantable tumor: implications for Marek’s
Avian Pathol. 16:51–60. vaccine mechanisms. Avian Dis. 55:293–301.
255 Hepkema, B.G, J.J Blankert, G.A Albers, M.G Tilanus, 268 Hunt, H.D, B. Lupiani, M.M Miller, I. Gimeno, L.F
E. Egberts, A.J van der Zijpp, and E.J Hensen. 1993. Lee, and M.S Parcells. 2001. Marek’s disease virus
Mapping of susceptibility to Marek’s disease within down‐regulates surface expression of MHC (B
the major histocompatibility (B) complex by refined Complex) Class I (BF) glycoproteins during active but
typing of White Leghorn chickens. Anim Genet. not latent infection of chicken cells. Virology.
24:283–287. 282:198–205.
256 Higgins, D.A, and B.W Calnek. 1975. Fowl 269 Ikezawa, M., M. Goryo, J. Sasaki, M. Haridy, and K.
immunoglobulins: quantitation in birds genetically Okada. Late 2010. Marek’s disease in adult chickens
resistant and susceptible to Marek’s disease. Infect inoculated with virulent Marek’s disease virus. J Vet
Immun. 12:360–363. Med Sci. 72:1539–1545.
257 Hildebrandt, E., J.R Dunn, and H.H Cheng. 2015. 270 Ikuta, K., H. Honma, K. Maotani, S. Ueda, S. Kato,
Addition of a UL5 helicase‐primase subunit point and K. Hirai. 1982. Monoclonal antibodies specific to
mutation eliminates bursal‐thymic atrophy of Marek’s and cross‐reactive with Marek’s disease virus and
disease virus Meq recombinant virus but reduces herpesvirus of turkeys. Biken J. 25:171–175.
vaccinal protection. Avian Pathol. 44:254–258. 271 Ikuta, K., K. Nakajima, M. Naito, S.H Ann, S. Ueda, S.
258 Hildebrandt, E., J.R Dunn, S. Perumbakkam, M. Kato, and K. Hirai. 1985. Identification of Marek’s
Niikura, and H.H Cheng. 2014. Characterizing the disease virus‐specific antigens in Marek’s disease
molecular basis of attenuation of Marek’s disease virus lymphoblastoid cell lines using monoclonal antibody
via in vitro serial passage identifies de novo mutations against virus‐specific phosphorylated polypeptides.
in the helicase‐primase subunit gene UL5 and other Int J Cancer. 35:257–264.
candidates associated with reduced virulence. J Virol. 272 Ikuta, K., K. Nakajima, S. Ueda, S. Kato, and K. Hirai.
88:6232–6242. 1985. Differences in the processing of secreted
259 Hirai, K., ed. 2001. Marek’s Disease. Springer‐Verlag, glycoprotein A induced by Marek’s disease virus and
Berlin. herpesvirus of turkeys. J Gen Virol. 66:1131–1137.
260 Hirai, K., K. Ikuta, and S. Kato. 1981. Structural 273 Isfort, R., H. Kung, and L. Velicer. 1987. Identification
changes of the DNA of Marek’s disease virus during of the gene encoding Marek’s disease herpesvirus A
serial passage in cultured cells. Virology. 115:385–389. antigen. J Virol. 61:2614–2620.
261 Hirai, K., and M. Sakaguchi. 2001. Polyvalent 274 Isfort, R.J, R.A Stringer, H.‐J. Kung, and L.F Velicer.
recombinant Marek’s disease virus vaccine against 1986. Synthesis, processing, and secretion of the
poultry diseases. Curr Top Microbiol Immunol. Marek’s disease herpesvirus A antigen glycoprotein.
255:261–287. J Virol. 57:464–474.
262 Hlozanek, I., V. Jurajda, and V. Benda. 1977. 275 Ishihara, Y., M. Esaki, S. Saitoh, T. Sato, and A.
Disinfection of Marek’s disease virus in poultry dust. Yasuda. 2016. Development and evaluation of the
Avian Pathol. 6:241–250. protective efficacy of novel Marek’s disease virus
263 Hlozanek, I., O. Mach, and V. Jurajda. 1973. Cell‐free rispens vector vaccines against infectious bursal
preprations of Marek’s disease virus from poultry disease. Avian Dis. 60:618–627.
dust. Folia Biol (Praha). 19:118–123. 276 Islam, A., B.F Cheetham, T.J Mahony, P.L Young, and
264 Holland, M.S, C.D Mackenzie, R.W Bull, and R.F S.W Walkden‐Brown. 2006. Absolute quantitation of
Silva. 1996. A comparative study of histological Marek’s disease virus and Herpesvirus of turkeys in
conditions suitable for both immunofluorescence and chicken lymphocyte, feather tip and dust samples
in situ hybridization in the detection of Herpesvirus using real‐time PCR. J Virol Methods. 132:127–134.
and its antigens in chicken tissues. J Histochem 277 Islam, A., B. Harrison, B.F Cheetham, T.J Mahony, P.L
Cytochem. 44:259–265. Young, and S.W Walkden‐Brown. 2004. Differential
265 Holland, M.S, C.D Mackenzie, R.W Bull, and R.F amplification and quantitation of Marek’s disease
Silva. 1998. Latent turkey herpesvirus infection in viruses using real‐time polymerase chain reaction.
lymphoid, nervous, and feather tissues of chickens. J Virol Methods. 119:103–113.
Avian Dis. 42:292–299. 278 Islam, A., and S.W Walkden‐Brown. 2007.
266 Hooft van Iddekinge, B.J, L. Stenzler, K.A Schat, H. Quantitative profiling of the shedding rate of the three
Boerrigter, and G. Koch. 1999. Genome analysis of Marek’s disease virus (MDV) serotypes reveals that
Marek’s disease virus strain CVI‐988: effect of cell challenge with virulent MDV markedly increases
culture passage on the inverted repeat regions. Avian shedding of vaccinal viruses. J Gen Virol.
Dis. 43:182–188. 88:2121–2128.
Chapter 15  Neoplastic Diseases 669

279 Islam, T., K.G Renz, S.W Walkden‐Brown, and S. responses after MDV infection. Avian Dis.
Ralapanawe. 2013. Viral kinetics, shedding profile, and 46:636–649.
transmission of serotype 1 Marek’s disease vaccine 291 Jeurissen, S.H, and G.F de Boer. 1993. Chicken
Rispens/CVI988 in maternal antibody‐free chickens. anaemia virus influences the pathogenesis of Marek’s
Avian Dis. 57:454–463. disease in experimental infections, depending on the
280 Islam, T., S.W Walkden‐Brown, K.G Renz, A.F Islam, dose of Marek’s disease virus. Vet Q. 15:81–84.
and S. Ralapanawe. 2014. Replication kinetics and 292 Johnson, D.I, A. Vagnozzi, F. Dorea, S.M Riblet, A.
shedding of very virulent Marek’s disease virus and Mundt, G. Zavala, and M. Garcia. 2010. Protection
vaccinal Rispens/CVI988 virus during single and against infectious laryngotracheitis by in ovo
mixed infections varying in order and interval vaccination with commercially available viral vector
between infections. Vet Microbiol. 173:208–223. recombinant vaccines. Avian Dis. 54:1251–1259.
281 Islam, T., S.W Walkden Brown, K.G Renz, A.F Fakhrul 293 Jordan, I., K. John, K. Howing, V. Lohr, Z. Penzes, E.
Islam, and S. Ralapanawe. 2013. Vaccination‐challenge Gubucz‐Sombor, Y. Fu, P. Gao, T. Harder, Z. Zadori,
interval markedly influences protection provided by and V. Sandig. 2016. Continuous cell lines from the
Rispens CVI988 vaccine against very virulent Marek’s Muscovy duck as potential replacement for primary
disease virus challenge. Avian Pathol. 42:516–526. cells in the production of avian vaccines. Avian
282 Izumiya, Y., H.K Jang, M. Ono, and T. Mikami. 2001. Pathol. 45:137–155.
A complete genomic DNA sequence of Marek’s 294 Jungherr, E., L.P Doyle, and E.P Johnson. 1941.
disease virus type 2, strain HPRS24. Curr Top Tentative pathologic nomenclature for the disease
Microbiol Immunol. 255:191–221. and/or for the disease complex variously designated
283 Jakowski, R.M, T.N Fredrickson, T.W Chomiak, and as fowl leukemia, fowl leucosis, etc. Am J Vet Res.
R.E Luginbuhl. 1970. Hematopoietic destruction in 2:116.
Marek’s disease. Avian Dis. 14:374–385. 295 Kaiser, P., G. Underwood, and F. Davison. 2003.
284 Jansen, C.A, P.M van de Haar, D. van Haarlem, P. van Differential cytokine responses following Marek’s
Kooten, t. S. de Wi, W. van Eden, B.C Viertlböck, T.W disease virus infection of chickens differing in
Göbel, and L. Vervelde. 2010. Identification of new resistance to Marek’s disease. J Virol. 77:762–768.
populations of chicken natural killer (NK) cells. Dev 296 Kamil, J., D. Robinson, L.F Lee, and H.J Kung. 2001.
Comp Immunol. 34:759–767. Marek’s disease virus encodes a secreted lipase. In:
285 Jarosinski, K., L. Kattenhorn, B. Kaufer, H. Ploegh, and Current Progress on Marek’s Disease Research. K.A
N. Osterrieder. 2007. A herpesvirus ubiquitin‐specific Schat, R.M Morgan, M.S Parcells and J.L Spencer, eds.
protease is critical for efficient T cell lymphoma American Association of Avian Pathologists, Kennett
formation. Proc Natl Acad Sci USA. Square, PA. 209–213.
104:20025–29930. 297 Kamil, J.P, B.K Tischer, S. Trapp, V.K Nair, N.
286 Jarosinski, K.W 2017. Interindividual Spread of Osterrieder, and H.J Kung. 2005. vLIP, a viral lipase
Herpesviruses. Advances in anatomy, embryology, homologue, is a virulence factor of Marek’s disease
and cell biology. Act Anat Embryol Cell Biol. virus. J Virol. 79:6984–6996.
223:195–224. 298 Kanamori, A., K. Ikuta, S. Ueda, S. Kato, and K. Hirai.
287 Jarosinski, K.W, S. Arndt, B.B Kaufer, and N. 1987. Methylation of Marek’s disease virus DNA in
Osterrieder. 2012. Fluorescently tagged pUL47 of chicken T‐ lymphoblastoid cell lines. J Gen Virol.
Marek’s disease virus reveals differential tissue 68:1485–1490.
expression of the tegument protein in vivo. J Virol. 299 Kaplan, S.K, and K.A Schat. 2001. Unpublished data.
86:2428–2436. 300 Kaschka‐Dierich, C., K. Nazerian, and R. Thomssen.
288 Jarosinski, K.W, B.L Njaa, P.H O’Connell, and K.A 1979. Intracellular state of Marek’s disease virus DNA
Schat. 2005. Pro‐inflammatory responses in chicken in two tumour‐derived chicken cell lines. J Gen Virol.
spleen and brain tissues after infection with very 44:271–280.
virulent plus Marek’s disease virus. Viral Immunol. 301 Kato, S., and K. Hirai. 1985. Marek’s disease virus. Adv
18:148–161. Virus Res. 30:225–277.
289 Jarosinski, K.W, N. Osterrieder, V.K Nair, and K.A 302 Kaufer, B.B 2013. Detection of integrated herpesvirus
Schat. 2005. Attenuation of Marek’s disease virus by genomes by fluorescence in situ hybridization (FISH).
deletion of open reading frame RLORF4 but not Methods Mol Biol. 1064:141–152.
RLORF5a. J Virol.. 79:11647–11659. 303 Kaufer, B.B, S. Arndt, S. Trapp, N. Osterrieder, and
290 Jarosinski, K.W, R.W Yunis, P.H O’Connell, C.J K.W Jarosinski. 2011. Herpesvirus telomerase RNA
Markowski‐Grimsrud, and K.A Schat. 2002. Influence (vTR) with a mutated template sequence abrogates
of genetic resistance of the chicken and virulence of herpesvirus‐induced lymphomagenesis. PLoS Pathog.
Marek’s disease virus (MDV) on nitric oxide 7:e1002333.
670 Section II  Viral Diseases

304 Kaufer, B.B, K.W Jarosinski, and N. Osterrieder. 2011. 317 Kodama, H., C. Sugimoto, F. Inage, and T. Mikami.
Herpesvirus telomeric repeats facilitate genomic 1979. Anti‐viral immunity against Marek’s disease
integration into host telomeres and mobilization of virus‐infected chicken kidney cells. Avian Pathol.
viral DNA during reactivation. J Exp Med. 8:33–44.
208:605–615. 318 Konobe, T., T. Ishikawa, K. Takaku, K. Ikuta, N.
305 Kaufer, B.B, S. Trapp, K.W Jarosinski, and N. Kitamoto, and S. Kato. 1979. Marek’s disease virus and
Osterrieder. 2010. Herpesvirus telomerase RNA(vTR)‐ herpesvirus of turkey noninfective to chickens,
dependent lymphoma formation does not require obtained by repeated in vitro passages. Biken J.
interaction of vTR with telomerase reverse 22:103–107.
transcriptase (TERT). PLoS Pathog. 6:e1001073. 319 Kreager, K. 1997. A global perspective on Marek’s
306 Kaul, L., and H.K Pradhan. 1991. Immunopathology of disease control in layers and layer breeders. World
Marek’s disease in quails: presence of antinuclear Poult.: S14–15.
antibody and immune complex. Vet Immunol 320 Kreager, K.S 1998. Chicken industry strategies for
Immunopathol. 28:89–96. control of tumor virus infections. Poult Sci.
307 Kawamura, H., J. King, D.J., and D.P Anderson. 1969. 77:1213–1216.
A herpesvirus isolated from kidney cell culture of 321 Kufuor‐Mensah, E., W.M Reed, S. Sleight, J.
normal turkeys. Avian Dis. 13:853–886. Pestka, A.M Fadly, and J.R Dunn. 2016. Effects of
308 Kaya, M., L. Preeyanon, J.B Dodgson, and H.H Cheng. T‐2 toxin on turkey herpesvirus‐induced vaccinal
2016. Validation of alternative transcript splicing in immunity against Marek’s disease. Avian Dis.
chicken lines that differ in genetic resistance to 60:56–62.
Marek’s disease. Anim Biotechnol. 27:238–244. 322 Kumar, M.A, R. Barathidasan, M. Palanivelu, S.D
309 Kennedy, D.A, J.R Dunn, P.A Dunn, and A.F Read. Singh, M.Y Wani, Y.S Malik, R. Singh, and K. Dhama.
2015. An observational study of the temporal and 2016. A novel recombinant Meq protein based dot‐
spatial patterns of Marek’s‐disease‐associated leukosis ELISA for rapid and confirmatory diagnosis of
condemnation of young chickens in the United States Marek’s disease induced lymphoma in poultry. J Virol
of America. Preventive veterinary medicine Methods. 236:271–280.
120:328–335. 323 Kumar, S., J.J Buza, and S.C Burgess. 2009. Genotype‐
310 Kenzy, S.G, and P.M Biggs. 1967. Excretion of the dependent tumor regression in Marek’s disease
Marek’s disease agent by infected chickens. Vet Rec. mediated at the level of tumor immunity. Cancer
80:565–568. Microenviron. 2:23–31.
311 Kenzy, S.G, B.R Cho, and Y. Kim. 1973. Oncogenic 324 Kung, H.J, L. Xia, P. Brunovskis, D. Li, J.L Liu, and L.F
Marek’s disease herpesvirus in avian encephalitis Lee. 2001. Meq: an MDV‐specific bZIP transactivator
(temporary paralysis). J Natl Cancer Inst. 51:977–982. with transforming properties. Curr Top Microbiol
312 Kheimar, A., R.L Previdelli, D.J Wight, and B.B Kaufer. Immunol. 255:245–260.
2017. Telomeres and telomerase: role in Marek’s 325 Lapen, R.F, and S.G Kenzy. 1972. Distribution of gross
disease virus pathogenesis, integration and cutaneous Marek’s disease lesions. Poult Sci.
tumorigenesis. Viruses. 9. 51:334–336.
313 Kim, T., J. Mays, A. Fadly, and R. Silva. 2011. 326 Lawn, A.M, and L.N Payne. 1979. Chronological study
Artificially inserting a reticuloendotheliosis virus long of ultrastructural changes in the peripheral nerves in
terminal repeat into a bacterial artificial chromosome Marek’s disease. Neuropathol Appl Neurobiol.
clone of Marek’s disease virus (MDV) alters expression 5:485–497.
of nearby MDV genes. Virus Genes. 42:369–376. 327 Lee, J.Y, D.N Foster, W.G Bottje, H.M Jang, Y.G
314 King, A.M.Q., M.J Adams, E.B Carstens, and E.J Chandra, L.E Gentles, and B.W Kong. 2013.
Lefkowitz, eds. 2011. Ninth Report of the International Establishment of an immortal chicken embryo liver‐
Committee on Taxonomy of Viruses. Elsevier derived cell line. Poultry Science. 92:1604–1612.
Academic Press, San Diego. 328 Lee, L.E, R.L Witter, S.M Reddy, P. Wu, N. Yanagida,
315 King, D., D. Page, K.A Schat, and B.W Calnek. 1981. and S. Yoshida. 2003. Protection and synergism by
Difference between influences of homologous and recombinant fowl pox vaccines expressing multiple
heterologous maternal antibodies on response to genes from Marek’s disease virus. Avian Dis.
serotype‐2 and serotype‐3 Marek’s disease vaccines. 47:549–558.
Avian Dis. 25:74–81. 329 Lee, L.F, X. Cui, Z. Cui, I. Gimeno, B. Lupiani, and
316 Kingham, B.F, V. Zelnik, J. Kopacek, V. Majerciak, E. S.M Reddy. 2005. Characterization of a very virulent
Ney, and C.J Schmidt. 2001. The genome of Marek’s disease virus mutant expressing the pp38
herpesvirus of turkeys: comparative analysis with protein from the serotype 1 vaccine strain CVI988/
Marek’s disease viruses. J Gen Virol. 82:1123–1135. Rispens. Virus Genes. 31:73–80.
Chapter 15  Neoplastic Diseases 671

330 Lee, L.F, K. Kreager, M. Heidari, H. Zhang, B. Lupiani, 341 Levy, A.M, O. Gilad, L. Xia, Y. Izumiya, J. Choi, A.
S.M Reddy, and A. Fadly. 2013. Properties of a Tsalenko, Z. Yakhini, R. Witter, L. Lee, C.J Cardona,
meq‐deleted rMd5 Marek’s disease vaccine: and H.J Kung. 2005. Marek’s disease virus Meq
protection against virulent MDV challenge and transforms chicken cells via the v‐Jun transcriptional
induction of lymphoid organ atrophy are cascade: A converging transforming pathway for avian
simultaneously attenuated by serial passage in vitro. oncoviruses. Proc Natl Acad Sci USA.
Avian Dis. 57:491–497. 102:14831–14836.
331 Lee, L.F, K.S Kreager, J. Arango, A. Paraguassu, B. 342 Levy, A.M, Y. Izumiya, P. Brunovskis, L. Xia, M.S
Beckman, H. Zhang, A. Fadly, B. Lupiani, and S.M Parcells, S.M Reddy, L.F Lee, H.W Chen, and H.J
Reddy. 2010. Comparative evaluation of vaccine Kung. 2003. Characterization of the chromosomal
efficacy of recombinant Marek’s disease virus vaccine binding sites and dimerization partners of the viral
lacking Meq oncogene in commercial chickens. oncoprotein Meq in Marek’s disease virus‐
Vaccine. 28:1294–1299. transformed T cells. J Virol. 77:12841–12851.
332 Lee, L.F, X. Liu, and R.L Witter. 1983. Monoclonal 343 Li, X., and K.A Schat. 2004. Quail cell lines supporting
antibodies with specificity for three different replication of Marek’s disease virus serotype 1 and 2
serotypes of Marek’s disease viruses in chickens. and herpesvirus of turkeys. Avian Dis. 48:803–812.
J Immunol. 130:1003–1006. 344 Li, Y., A. Sun, S. Su, P. Zhao, Z. Cui, and H. Zhu. 2011.
333 Lee, L.F, B. Lupiani, R.F Silva, H.‐J. Kung, and S.M Deletion of the Meq gene significantly decreases
Reddy. 2008. Recombinant Marek’s disease virus immunosuppression in chickens caused by pathogenic
(MDV) lacking the Meq oncogene confers protection Marek’s disease virus. Virol J. 8:2.
against challenge with a very virulent plus strain of 345 Lin, J.A, H. Kodama, M. Onuma, and T. Mikami.
MDV. Vaccine. 26:1887–1892. 1991. The early pathogenesis in chicken inoculated
334 Lee, L.F, J.M Sharma, K. Nazerian, and R.L Witter. with non‐pathogenic serotype 2 Marek’s disease virus.
1978. Suppression of mitogen‐induced proliferation of J Vet Med Sci. 53:269–273.
normal spleen cells by macrophages from chickens 346 Liu, H.C, H.J Kung, J.E Fulton, R.W Morgan, and H.H
inoculated with Marek’s disease virus. J Immunol. Cheng. 2001. Growth hormone interacts with the
120:1554–1559. Marek’s disease virus SORF2 protein and is associated
335 Lee, L.F, P. Wu, D. Sui, D. Ren, J. Kamil, H.J Kung, and with disease resistance in chicken. Proc Natl Acad Sci
R.L Witter. 2000. The complete unique long sequence USA. 98:9203–9208.
and the overall genomic organization of the GA strain 347 Liu HC, N.M, J.E Fulton, H.H Cheng. 2003.
of Marek’s disease virus. Proc Natl Acad Sci USA. Identification of chicken lymphocyte antigen 6
97:6091–6096. complex, locus E (LY6E, alias SCA2) as a putative
336 Lee, L.F, H. Zhang, M. Heidari, B. Lupiani, and S.M Marek’s disease resistance gene via a virus‐host
Reddy. 2011. Evaluation of factors affecting vaccine protein interaction screen. Cytogenetic Genome Res.
efficacy of recombinant Marek’s disease virus lacking 102:304–308.
the Meq oncogene in chickens. Avian Dis. 55:172–179. 348 Liu, H.C, E.J Soderblom, and M.B Goshe. 2006. A
337 Lee, S.I, K. Ohashi, T. Morimura, C. Sugimoto, and M. mass spectrometry‐based proteomic approach to
Onuma. 1999. Re‐isolation of Marek’s disease virus study Marek’s disease virus gene expression. J Virol
from T cell subsets of vaccinated and non‐vaccinated Methods. 135:66–75.
chickens. Arch Virol. 144:45–54. 349 Liu, Y., K. Li, Y. Gao, L. Gao, L. Zhong, Y. Zhang, C.
338 Lee, S.I, K. Ohashi, C. Sugimoto, and M. Onuma. Liu, Y. Zhang, and X. Wang. Recombinant Marek’s
2001. Heparin inhibits plaque formation by cell‐free Disease 2016. Virus as a vector‐based vaccine against
Marek’s disease viruses in vitro. J Vet Med Sci. avian leukosis virus subgroup J in chicken. Viruses. 8.
63:427–432. 350 Luo, J., Y. Yu, S. Chang, F. Tian, H. Zhang, and J. Song.
339 Lemiere, S., S.Y Wong, A.L Saint‐Gerand, S. 2012. DNA methylation fluctuation induced by virus
Goutebroze, and F.X Le Gros. 2011. Compatibility of infection differs between MD‐resistant and ‐
turkey herpesvirus‐infectious bursal disease vector susceptible chickens. Front Genet. 3:20.
vaccine with Marek’s disease rispens vaccine injected 351 Luo, J., Y. Yu, H.M Zhang, F. Tian, S. Chang, H.H
into day‐old pullets. Avian Dis. 55:113–118. Cheng, and J. Song. 2011. Down‐regulation of
340 Levy, A.M, S.C Burgess, I. Davidson, G. Underwood, promoter methylation level of CD4 gene after MDV
G. Leitner, and E.D Heller. 2003. Interferon‐ infection in MD‐susceptible chicken line. BMC Proc.
containing supenatants increase Marek’s disease 5(4):S7.
herpesvirus genomes and gene transcription levels, 352 Lupiani, B., L.F Lee, X. Cui, I. Gimeno, A. Anderson,
but not virion replication in vitro. Viral Immunol. R.F Silva, R.L Witter, H.J Kung, and S.M Reddy. 2004.
16:501–509. Marek’s disease virus‐encoded Meq gene is involved
672 Section II  Viral Diseases

in transformation of lymphocytes but is dispensable virulent Marek’s disease virus containing a


for replication. Proc Natl Acad Sci USA. reticuloendothelial virus long terminal repeat. Avian
101:11815–11820. Pathol. 1–31.
353 Lupiani, B., L.F Lee, K.S Kreager, R.L Witter, and S.M 365 McColl, K., B.W Calnek, W.V Harris, K.A Schat, and
Reddy. 2013. Insertion of reticuloendotheliosis virus L.F Lee. 1987. Expression of a putative tumor‐
long terminal repeat into the genome of CVI988 strain associated antigen on normal versus Marek’s disease
of Marek’s disease virus results in enhanced growth virus‐transformed lymphocytes. J Natl Cancer Inst.
and protection. Avian Dis. 57:427–431. 79:991–1000.
354 Lupiani, B., L.F Lee, and S.M Reddy. 2001. Protein‐ 366 McKay, J.C 1998. A poultry breeder’s approach to
coding content of the sequence of Marek’s disease avian neoplasia. Avian Pathol. 27:S74–S77.
virus serotype 1. Curr Top Microbiol Immunol. 367 McPherson, M.C, and M.E Delany. 2016. Virus and
255:159–190. host genomic, molecular, and cellular interactions
355 Maas, H.J L., H.W Antonisse, V.D A.J Zypp, J.E during Marek’s disease pathogenesis and oncogenesis.
Groenendal, and G.L Kok. 1981. The development of Poult Sci. 95:412–429.
two white plymouth rock lines resistant to Marek’s 368 Mete, A., R. Gharpure, M.E Pitesky, D. Famini, K.
disease by breeding from survivors. Avian Pathol. Sverlow, and J. Dunn. 2016. Marek’s disease in
10:137–115. backyard chickens, a study of pathologic findings and
356 Maas, H.J L., B.H Rispens, and J.E Groenendal. 1974. viral loads in tumorous and nontumorous birds. Avian
Control of Marek’s disease in the Netherlands: large Dis. 60:826–836.
scale field trials with the avirulent cell‐associated 369 Mete, A., F. Giannitti, B. Barr, L. Woods, and M.
Marek’s disease vaccine virus (strain CVI988). Anderson. 2013. Causes of mortality in backyard
Tijdschr Diergeneeskd. 99:1273–1288. chickens in northern California: 2007–2011. Avian
357 Maas, H.J L., A.J van der Zijpp, J.E Groenendal, and Dis. 57:311–315.
G.L Kok. 1982. Evaluation of resistance to Marek’s 370 Miles, A.M, A.S Anderson, E.L Bernberg, J. Kent, J.K
disease in three white plymouth rock lines and in two Rosenberger, C.R Pope, and R.W Morgan. 1999.
reciprocal crosses. Avian Pathol. 11:1–9. Comparison of two serotype 1 MDV isolates. Acta
358 MacEachern, S., W.M Muir, S. Crosby, and H.H Virol. 43:102–105.
Cheng. 2011. Genome‐wide identification of allele‐ 371 Miles, A.M, S.M Reddy, and R.W Morgan. 2001.
specific expression (ASE) in response to Marek’s Coinfection of specific‐pathogen‐free chickens with
disease virus infection using next generation Marek’s disease virus (MDV) and chicken infectious
sequencing. BMC Proc. 5:S14. anemia virus: effect of MDV pathotype. Avian Dis.
359 Maotani, K., A. Kanamori, K. Ikuta, S. Ueda, S. Kato, 45:9–18.
and K. Hirai. 1986. Amplification of a tandem direct 372 Minick, C.R, C.G Fabricant, J. Fabricant, and M.M
repeat within inverted repeats of Marek’s disease virus Litrenta. 1979. Atheroarteriosclerosis induced by
DNA during serial in vitro passage. J Virol. infection with a herpesvirus. Am J Pathol.
58:657–660. 96:673–706.
360 Marek, J. 1907. Multiple Nervenentzuendung 373 Mohammadi, A., J.L Spencer, M. Chan, and M. Ansari
(Polyneuritis) bei Huehnern. Dtsch. Tierarztl Lari. 2007. Antibody response of chickens to serotype
Wochenschr. 15:417–421. 1, 2, or 3 Marek’s disease vaccines based on ELISA
361 Markowski‐Grimsrud, C.J, and K.A Schat. Cytotoxic with infected cells as antigen. Avian Dis. 51:982–985.
2002. T lymphocyte responses to Marek’s disease 374 Morgan, R.W, J. Gelb, Jr, C.S Schreurs, D. Lutticken,
herpesvirus‐encoded glycoproteins. Vet Immunol J.K Rosenberger, and P.J A. Sondermeijer. 1992.
Immunopathol. 90:133–144. Protection of chickens from Newcastle and Marek’s
362 Markowski‐Grimsrud, C.J, and K.A Schat. 2003. diseases with a recombinant herpesvirus of turkeys
Infection with chicken anemia virus impairs the vaccine expressing the Newcastle disease virus fusion
generation of antigen‐specific cytotoxic T protein. Avian Dis. 36:858–870.
lymphocytes. Immunol. 109:283–294. 375 Morgan, R.W, L. Sofer, A.S Anderson, E.L Bernberg, J.
363 Martin, A., E.A Dunnington, W.E Briles, R.W Briles, Cui, and J. Burnside. 2001. Induction of host gene
and P.B Siegel. 1989. Marek’s disease and major expression following infection of chicken embryo
histocompatibility complex haplotypes in chickens fibroblasts with oncogenic Marek’s disease virus.
selected for high or low antibody response. Anim J Virol. 75:533–539.
Genet. 20:407–414. 376 Morgan, R.W, Q. Xie, J.L Cantello, A.M Miles, E.L
364 Mays, J.K, A. Black‐Pyrkosz, S. Spatz, A.M Fadly, and Bernberg, J. Kent, and A. Anderson. 2001. Marek’s
J.R Dunn. 2016. Protective efficacy of a recombinant disease virus latency. Curr Top Microbiol Immunol.
bacterial artificial chromosome clone of a very 255:223–243.
Chapter 15  Neoplastic Diseases 673

377 Moriguchi, R., M. Oshima, F. Mori, I. Umezawa, and Disease: An Evolving Problem. F. Davison and V. Nair,
C. Itakura. 1989. Chronological change of feather pulp eds. Elsevier Academic Press, London. 32–48.
lesions during the course of Marek’s disease virus‐ 389 Nair, V., and M. Zavolan. 2006. Virus‐encoded
induced lymphoma formation in field chickens. In: microRNAs: novel regulators of gene expression.
Advances in Marek’s Disease Research. S. Kato, T. Trends Microbiol. 14:169–175.
Horiuchi, T. Mikami and K. Hirai, eds. Japanese 390 Naito, M., K. Nakajima, N. Iwa, K. Ono, I. Yoshida, T.
Association on Marek’s Disease, Osaka, Japan. Konobe, K. Ikuta, S. Ueda, S. Kato, and K. Hirai. 1986.
338–343. Demonstration of a Marek’s disease virus‐specific
378 Morimura, T., M. Hattori, K. Ohashi, C. Sugimoto, antigen in tumour lesions of chickens with Marek’s
and M. Onuma. 1995. Immunomodulation of disease using monoclonal antibody against a virus
peripheral T cells in chickens infected with Marek’s phosphorylated protein. Avian Pathol. 15:503–510.
disease virus: involvement in immunosuppression. 391 Nakamura, K., M. Ito, K. Fujino, Y. Yamamoto, M.
J Gen Virol. 79:2979–2985. Mase, M. Yamada, H. Kobayashi, and T. Harada. 2010.
379 Morimura, T., K. Ohashi, Y. Kon, M. Hattori, C. Pathology and microbiology of dermal squamous cell
Sugimoto, and M. Onuma. Apoptosis and 1996. carcinoma in young brown chickens reared on reused
CD8‐down‐regulation in the thymus of chickens litter. Avian Dis. 54:1120–1124.
infected with Marek’s disease virus. Arch Virol. 392 Nazerian, K. 1987. An updated list of avian cell lines
141:2243–2249. and transplantable tumours. Avian Pathol. 16:527–544.
380 Morimura, T., K. Ohashi, Y. Kon, M. Hattori, C. 393 Nazerian, K., L.F Lee, R.L Witter, and B.R Burmester.
Sugimoto, and M. Onuma. 1997. Apoptosis in 1970. Ultra‐structural studies of a herpesvirus of
peripheral CD4+T cells and thymocytes by Marek’s turkeys antigenically related to Marek’s disease virus.
disease virus‐infection. Leukemia 11 Suppl 3:206–208. Virology. 43:442–452.
381 Morrow, C., and F. Fehler. 2004. Marek’s disease: a 394 Nazerian, K., L.F Lee, N. Yanagida, and R. Ogawa.
worldwide problem. In: Marek’s disease. An evolving 1992. Protection against Marek’s disease by a fowlpox
problem. F. Davison and V. Nair, eds. Elsevier virus recombinant expressing the glycoprotein B of
Academic Press, London. 49–61. Marek’s disease virus. J Virol. 66:1409–1413.
382 Murata, S., K.‐S. Chang, S.‐I. Lee, S. Konnai, M. 395 Nazerian, K., R.L Witter, L.F Lee, and N. Yanagida.
Onuma, and K. Ohashi. 2007. Development of a 1996. Protection and synergism by recombinant fowl
nested polymerase chain reaction method to detect pox vaccines expressing genes from Marek’s disease
oncogenic Marek’s disease virus from feather tips. virus. Avian Dis. 40:368–376.
J Vet Diagn Invest. 19:471–478. 396 Niikura, M., T. Kim, H.D Hunt, J. Burnside, R.W
383 Murata, S., K.S Chang, Y. Yamamoto, T. Okada, S.I Morgan, J.B Dodgson, and H.H Cheng. 2007. Marek’s
Lee, S. Konnai, M. Onuma, Y. Osa, M. Asakawa, and disease virus up‐regulates major histocompatibility
K. Ohashi. 2007. Detection of the virulent Marek’s complex class II cell surface expression in infected
disease virus genome from feather tips of wild geese cells. Virology. 359:212–219.
in Japan and the Far East region of Russia. Arch Virol. 397 Niikura, M., H.‐C. Liu, J.B Dodgson, and H.H Cheng.
152. 2004. A comprehensive screen for chicken proteins
384 Murata, S., Y. Hayashi, A. Kato, M. Isezaki, S. that interact with proteins unique to virulent strains
Takasaki, M. Onuma, Y. Osa, M. Asakawa, S. Konnai, of Marek’s disease virus. Poult Sci. 83:1117–1123.
and K. Ohashi. 2012. Surveillance of Marek’s disease 398 Niikura, M., R.L Witter, H.K Jang, M. Ono, T. Mikami,
virus in migratory and sedentary birds in Hokkaido. and R.F Silva. 1999. MDV glycoprotein D is expressed
Japan Vet J. 192:538–540. in the feather follicle epithelium of infected chickens.
385 Murata, S., T. Okada, R. Kano, Y. Hayashi, T. Acta Virol. 43:159–163.
Hashiguchi, M. Onuma, S. Konnai, and K. Ohashi. 399 Njenga, M.K, and C.A Dangler. 1996. Intimal lipid
2011. Analysis of transcriptional activities of the Meq accretion and elevated serum cholesterol in Marek’s
proteins present in highly virulent Marek’s disease disease virus‐inoculated chickens. Vet Pathol.
virus strains, RB1B and Md5. Virus Genes. 43:66–71. 33:704–708.
386 Mwangi, W.N, L.P Smith, S.J Baigent, R.K Beal, V. 400 Ohashi, K., W. Zhou, P.H O’Connell, and K.A Schat.
Nair, and A.L Smith. 2011. Clonal structure of rapid‐ 1994. Characterization of a Marek’s disease virus
onset MDV‐driven CD4+ lymphomas and responding BamHI‐L specific cDNa clone obtained from a
CD8+ T cells. PLoS Pathog. 7:e1001337. Marek’s disease lymphoblastoid cell line. J Virol.
387 Nair, V. 2013. Latency and Tumorigenesis in Marek’s 68:1191–1195.
Disease. Avian Dis. 57:360–365. 401 OIE. 2010. Marek’s disease Manual of Diagnostic
388 Nair, V., and H.J Kung. 2004. Marek’s disease virus Tests and Vaccines for Terrestrial Animals. Manual of
oncogenicity: molecular mechanisms. In: Marek’s Diagnostic Tests and Vaccines for Terrestrial Animals.
674 Section II  Viral Diseases

402 Okada, K., Y. Tanaka, K. Murakami, S. Chiba, T. 414 Pandiri, A.K, A.L Cortes, L.F Lee, and I.M Gimeno.
Morimura, M. Hattori, M. Goryo, and M. Onuma. 2008. Marek’s disease virus infection in the eye:
1997. Phenotype analysis of lymphoid cells in Marek’s chronological study of the lesions, virus replication,
disease of CD4+ or CD8+ T‐cell‐deficient chickens: and vaccine‐induced protection. Avian Dis.
occurrence of double negative T‐cell turnout. Avian 52:572–580.
Pathol. 26:525–534. 415 Pappenheimer, A.M, L.C Dunn, and V. Cone. 1929.
403 Okazaki, W., H.G Purchase, and B.R Burmester. Studies on fowl paralysis (neurolymphomatosis
1970. Protection against Marek’s disease by gallinarum). I. Clinical features and pathology. J Exp
vaccination with a herpesvirus of turkeys. Avian Dis. Med. 49:63–86.
14:413–429. 416 Parcells, M., J. Burnside, and R. Morgan. 2012.
404 Omar, A.R, and K.A Schat. 1996. Syngeneic Marek’s Marek’s disease virus‐induced T‐cell lymphomas. In:
disease virus (MDV)‐specific cell‐mediated immune Cancer Associated Viruses‐Current Research. E.S
responses against immediate early, late, and unique Robertson, ed. Springer Science+Business Media.
MDV proteins. Virology. 222:87–99. 307–335.
405 Omar, A.R, and K.A Schat. 1997. Characterization of 417 Parcells, M.S, R.L Dienglewicz, A.S Anderson, and
Marek’s disease herpesvirus‐specific cytotoxic T R.W Morgan. 1999. Recombinant Marek’s disease
lymphocytes in chickens inoculated with a non‐ virus (MDV)‐derived lymphoblastoid cell lines:
oncogenic vaccine strain of MDV. Immunol. regulation of a marker gene within the context of the
90:579–585. MDV genome. J Virol. 73:1362–1373.
406 Ono, M., H.K Jang, K. Maeda, Y. Kawaguchi, Y. Tohya, 418 Parker, M.A, and L.W Schierman. 1983. Suppression
M. Niikura, and T. Mikami. 1996. Detection of of humoral immunity in chickens prevents transient
Marek’s disease virus serotype 1 (MDV1) glycoprotein paralysis caused by a herpesvirus. J Immunol.
D in MDV1‐infected chick embryo fibroblasts. J Vet 130:2000–2001.
Med Sci. 58:777–780. 419 Parvizi, P., M.F Abdul‐Careem, A.I Mallick, K. Haq,
407 Ono, M., Y. Kawaguchi, K. Maeda, N. Kamiya, Y. H.R Haghighi, S. Orouji, M. Heidari, S. Behboudi, and
Tohya, C. Kai, M. Niikura, and T. Mikami. 1994. S. Sharif. 2014. The effects of administration of
Nucleotide sequence analysis of Marek’s disease virus ligands for Toll‐like receptor 4 and 21 against Marek’s
(MDV) serotype 2 homolog of MDV serotype 1 pp38, disease in chickens. Vaccine. 32:1932–1938.
an antigen associated with transformed cells. Virology. 420 Parvizi, P., K. Andrzejewski, L.R Read, S. Behboudi,
201:142–146. and S. Sharif. 2010. Expression profiling of genes
408 Osterrieder, N., J.P Kamil, D. Schumacher, B.K associated with regulatory functions of T‐cell subsets
Tischer, and S. Trapp. 2006. Marek’s disease virus: in Marek’s disease virus‐infected chickens. Avian
from miasma to model. Nat Rev Microbiol. 4:283–294. Pathol. 39:367–373.
409 Ottiger, H.P 2010. Development, standardization and 421 Parvizi, P., A.I Mallick, K. Haq, H.R Haghighi, S.
assessment of PCR systems for purity testing of avian Orouji, N. Thanthrige‐Don, M. St Paul, J.T Brisbin,
viral vaccines. Biologicals. 38:381–388. L.R Read, S. Behboudi, and S. Sharif. 2012. A toll‐like
410 Padhi, A., and M.S Parcells. Positive Selection Drives receptor 3 ligand enhances protective effects of
Rapid 2016. Evolution of the meq oncogene of Marek’s vaccination against Marek’s disease virus and hinders
disease virus. PLoS One. 11:e0162180. tumor development in chickens. Viral Immunol.
411 Palya, V., I. Kiss, T. Tatar‐Kis, T. Mato, B. Felfoldi, and 25:394–401.
Y. Gardin. 2012. Advancement in vaccination against 422 Parvizi, P., L.R Read, M.F Abdul‐Careem, C. Lusty,
Newcastle disease: recombinant HVT NDV provides and S. Sharif. 2009. Cytokine gene expression in
high clinical protection and reduces challenge virus splenic CD4(+) and CD8(+) T‐cell subsets of chickens
shedding with the absence of vaccine reactions. Avian infected with Marek’s disease virus. Viral Immunol.
Dis. 56:282–287. 22:31–38.
412 Palya, V., E.W Kovacs, T. Tatar‐Kis, B. Felfoldi, Z.G 423 Pattison, M. 1985. Control of Marek’s disease by the
Homonnay, T. Mato, T. Sato, and Y. Gardin. 2016. poultry industry: Practical considerations. In: Marek’s
Recombinant turkey herpesvirus‐AI vaccine virus Disease. L.N Payne, ed. Martinus Nijhoff, Boston,
replication in different species of waterfowl. Avian MA. 341–349.
Dis. 60:210–217. 424 Payne, L.N. 1985. Pathology. In: Marek’s Disease. L.N.
413 Pandey, U., A.S Bell, D.W Renner, D. Kennedy, J.T Payne, ed. Martinus Nijhoff, Boston, MA. 43–75.
Shreve, C. Cairns, M. Jones, P. Dunn, A.F Read, and 425 Payne, L.N 2004. Pathological responses to infection.
M.L Szpara. 2016. DNA from dust: the first field‐ In: Marek’s Disease: An Evolving Problem. T.F Davison
isolated genomes of MDV‐1, from virions in poultry and V.K Nair, eds. Elsevier Academic Press, London.
dust and chicken feather follicles. bioRxiv. 78–97.
Chapter 15  Neoplastic Diseases 675

426 Payne, L.N, and P.M Biggs. 1967. Studies on Marek’s Questionnaire study and postmortem findings in
disease. II. Pathogenesis. J Natl Cancer Inst. backyard chicken flocks in Finland. Acta Vet Scand. 57.
39:281–302. 439 Pol, J.M, G.L Kok, H.L Oei, and G.F de Boer. 1986.
427 Payne, L.N, J.A Frazier, and P.C Powell. 1976. Pathogenicity studies with plaque‐purified
Pathogenesis of Marek’s disease. Int Rev Exp Pathol. preparations of Marek’s disease virus strain CVI‐988.
16:59–154. Avian Dis. 30:271–275.
428 Payne, L.N, K. Howes, M. Rennie, J.M Bumstead, and 440 Pol, J.M A., G.L Kok, and G.F de Boer. 1985. Studies
A.W Kidd. 1981. Use of an agar culture technique for on the oncogenic properties of various Marek’s
establishing lymphoid cell lines from Marek’s disease disease virus strains. In: Proc Int Symp Marek’s Dis.
lymphomas. Int J Cancer. 28:757–766. B.W Calnek and J.L Spencer, eds. American
429 Payne, L.N, and M. Rennie. 1973. Pathogenesis of Association of Avian Pathologists, Kennett Square,
Marek’s disease in chicks with and without maternal PA. 469–479.
antibody. J Natl Cancer Inst. 51:1559–1573. 441 Powell, P.C, and T.F Davison. 1986. Induction of
430 Payne, L.N, and M. Rennie. 1976. Sequential changes Marek’s disease in vaccinated chickens by treatment
in the numbers of B and T lymphocytes and other with betamethasone or corticosterone. Israel J Vet
leukocytes in the blood in Marek’s disease. Int J Med. 42:73–78.
Cancer. 18:510–520. 442 Powell, P.C, K.J Hartley, B.M Mustill, and M. Rennie.
431 Payne, L.N, and J. Roszkowski. 1972. The presence of 1983. The occurrence of chicken foetal antigen after
immunologically uncommitted bursa and thymus infection with Marek’s disease virus in three strains of
dependent lymphoid cells in the lymphomas of chicken. Oncodev Biol Med. 4:261–271.
Marek’s disease. Avian Pathol. 1:27–34. 443 Powell, P.C, L.N Payne, J.A Frazier, and M. Rennie.
432 Peng, Q., and Y. Shirazi. 1996. Characterization of the 1974. T lymphoblastoid cell lines from Marek’s disease
protein product encoded by a splicing variant of the lymphomas. Nature. 251:79–80.
Marek’s disease virus Eco‐Q gene (Meq). Virology. 444 Pradhan, H.K, G.C Mohanty, W.Y Lee, L. Kaul, and
226:77–82. J.M Kataria. 1988. Immune complex‐mediated
433 Peng, Q., and Y. Shirazi. 1996. Isolation and glomerulopathy in Marek’s disease. Vet Immunol
characterization of Marek’s disease virus (MDV) Immunopathol. 19:165–171.
cDNAs from a MDV‐transformed lymphoblastoid cell 445 Prasad, L.B M., and P.B Spradbrow. 1977.
line: identification of an open reading frame antisense Multiplication of turkey herpes virus and Marek’s
to the MDV Eco‐Q protein (Meq). Virology. 221:368–374. disease virus in chick embryo skin cell cultures.
434 Pepose, J.S, J.G Stevens, M.L Cook, and P.W Lampert. J Comp Path. 87:515–520.
1981. Marek’s disease as a model for the Landry‐ 446 Prasad, L.M B., and P.B Spradbrow. 1980.
Guillain‐Barr‚ Syndrome: Latent viral infection in Ultrastructure and infectivity of tissue from normal
nonneuronal cells is accompanied by specific immune and immunodepressed chickens inoculated with
responses to peripheral nerve and myelin. Am J turkey herpesvirus. J Comp Pathol. 90:47–56.
Pathol. 103:309–332. 447 Pratt, W.D, J. Cantello, R.W Morgan, and K.A
435 Perozo, F., A.P Villegas, R. Fernandez, J. Cruz, and N. Schat. 1994. Enhanced expression of the Marek’s
Pritchard. 2009. Efficacy of single dose recombinant disease virus‐specific phosphoproteins after stable
herpesvirus of turkey infectious bursal disease virus transfection of MSB‐1 cells with the Marek’s
(IBDV) vaccination against a variant IBDV strain. disease virus homolog of ICP4. Virology.
Avian Dis. 53:624–628. 201:132–136.
436 Perumbakkam, S., W.M Muir, A. Black‐Pyrkosz, R. 448 Pratt, W.D, R. Morgan, and K.A Schat. 1992. Cell‐
Okimoto, and H.H Cheng. 2013. Comparison and mediated cytolysis of lymphoblastoid cells expressing
contrast of genes and biological pathways responding Marek’s disease virus‐specific phosphoproteins. Vet
to Marek’s disease virus infection using allele‐specific Microbiol. 33:93–99.
expression and differential expression in broiler and 449 Prineas, J.W, and R.G Wright. 1972. The fine structure
layer chickens. BMC Genomics. 14:64. of peripheral nerve lesions in a virus‐induced
437 Petherbridge, L., H. Xu, Y. Zhao, L.P Smith, J. demyelinating disease in fowl (Marek’s disease). Lab
Simpson, S. Baigent, and V. Nair. 2009. Cloning of Invest. 26:548–557.
Gallid herpesvirus 3 (Marek’s disease virus 450 Proudfoot, F.G, and J.R Aitken. 1969. The effect of diet
serotype‐2) genome as infectious bacterial artificial on mortality attributed to Marek’s disease among
chromosomes for analysis of viral gene functions. leghorn genotype. Poult Sci. 48:1457—1459.
J Virol Methods. 158:11–17. 451 Purchase, H.G 1972. Recent advances in the
438 Pohjola, L., L. Rossow, A. Huovilainen, T. Soveri, M.L knowledge of Marek’s disease. Adv Vet Sci Comp Med.
Hanninen, and M. Fredriksson‐Ahomaa. 2015. 16:223–258.
676 Section II  Viral Diseases

452 Purchase, H.G 1985. Clinical disease and its economic 465 Ren, D., L.F Lee, and P.M Coussens. 1994.
impact. In: Marek’s Disease. L.N Payne, ed. Martinus Identification and characterization of Marek’s disease
Nijhoff, Boston, MA. 17–24. virus genes homologous to ICP27 and glycoprotein K
453 Purchase, H.G, B.R Burmester, and C.H Cunningham. of herpes simplex virus‐1. Virology. 204:242–250.
1971. Responses of cell cultures from various avian 466 Ren, D., L.F Lee, and P.M Coussens. 1996. Regulatory
species to Marek’s disease virus and herpesvirus of function of the Marek’s disease virus ICP27 gene
turkeys. Am J Vet Res. 32:1811–1823. product. In: Current Research on Marek’s Disease. R.F
454 Purchase, H.G, W. Okazaki, and B.R Burmester. 1972. Silva, H.H Cheng, P.M Coussens, L.F Lee and L.F
Long‐term field trials with the herpesvirus of turkeys Velicer, eds. American Association of Avian
vaccine against Marek’s disease. Avian Dis. 16:57–71. Pathologists, Kennett Square, PA. 170–175.
455 Purchase, H.G, and R.L Witter. 1986. Public health 467 Rennie, M., P.C Powell, and B.M Mustill. 1980. The
concerns from human exposure to oncogenic avian effect of bursectomy on vaccination against Marek’s
herpesviruses. J Am Vet Med Assoc. 189:1430–1436. disease with the herpesvirus of turkeys. Avian Pathol.
456 Quéré, P. 1992. Suppression mediated in vitro by 9:557–566.
Marek’s disease virus‐transformed T‐lymphoblastoid 468 Renz, K.G, B.F Cheetham, and S.W Walkden‐Brown.
cell lines: effect on lymphoproliferation. Vet Immunol 2013. Differentiation between pathogenic serotype 1
Immunopathol. 32:149–164. isolates of Marek’s disease virus and the Rispens
457 Ralapanawe, S., K.G Renz, S.K Burgess, and S.W CVI988 vaccine in Australia using real‐time PCR and
Walkden‐Brown. 2016. Field studies of the detection, high resolution melt curve analysis. J Virol Methods.
persistence and spread of the Rispens CVI988 vaccine 187:144–152.
virus and the extent of co‐infection with Marek’s 469 Renz, K.G, A. Islam, B.F Cheetham, and S.W
disease virus. Aust Vet J. 94:329–337. Walkden‐Brown. 2006. Absolute quantification using
458 Ralapanawe, S., S.W Walkden‐Brown, A.F Islam, and real‐time polymerase chain reaction of Marek’s
K.G Renz. 2016. Effects of Rispens CVI988 disease virus serotype 2 in field dust samples, feather
vaccination followed by challenge with Marek’s tips and spleens. J Virol Methods. 135:186–191.
disease viruses of differing virulence on the 470 Rispens, B.H, H. van Vloten, N. Mastenbroek, H.J
replication kinetics and shedding of the vaccine and Maas, and K.A Schat. 1972. Control of Marek’s disease
challenge viruses. Vet Microbiol. 183:21–29. in the Netherlands. I. Isolation of an avirulent Marek’s
459 Ralapanawe, S., S.W Walkden‐Brown, K.G Renz, and disease virus (strain CVI 988) and its use in laboratory
A.F Islam. 2016. Protection provided by Rispens vaccination trials. Avian Dis. 16:108–125.
CVI988 vaccine against Marek’s disease virus isolates 471 Rong, S., D. Wheeler, and F. Weber. 2014. Efficient
of different pathotypes and early prediction of vaccine Marek’s disease virus (MDV) and herpesvirus of
take and MD outcome. Avian Pathol. 45:26–37. turkey infection of the QM7 cell line that does not
460 Ramachandra, R.N, R. Raghavan, and B.S Keshavamurthy. contain latent MDV genome. Avian Pathol. 43:414–419.
1978. Propagation of Marek’s disease virus in chicken 472 Ross, L.J N. 1980. Mechanism of protection conferred
tracheal explants. Indian J Anim Sci. 48:525–528. by HVT. In: Resistance and Immunity to Marek’s
461 Read, A.F, S.J Baigent, C. Powers, L.B Kgosana, L. Disease. P.M Biggs, ed. Commission of the European
Blackwell, L.P Smith, D.A Kennedy, S.W Walkden‐ Communities, Luxembourg. 289–297.
Brown, and V.K Nair. 2015. Imperfect vaccination can 473 Ross, L.J N. 1985. Molecular biology of the virus. In:
enhance the transmission of highly virulent Marek’s Disease. L.N Payne, ed. Martinus Nijhoff,
pathogens. PLoS Biol. 13:e1002198. Boston, MA. 113–150.
462 Reddy, S.M, Y. Izumiya, and B. Lupiani. 2016. Marek’s 474 Ross, L.J N., M.M Binns, P. Tyers, J. Pastorek, V.
disease vaccines: Current status, and strategies for Zelnik, and S.D Scott. 1993. Construction and
improvement and development of vector vaccines. Vet properties of a turkey herpesvirus recombinant
Microbiol. expressing the Marek’s disease virus homologue of
463 Reddy, S.M, B. Lupiani, I.M Gimeno, R.F Silva, L.F glycoprotein B of herpes simplex virus. J Gen Virol.
Lee, and R.L Witter. 2002. Rescue of a pathogenic 74:371–377.
Marek’s disease virus with overlapping cosmid DNAs: 475 Ross, L.J N., W. Delorbe, H.E Varmus, J.M Bishop, and
use of a pp38 mutant to validate the technology for M. Brahic. 1981. Persistence and expression of
the study of gene function. Proc Natl Acad Sci USA. Marek’s disease virus DNA in tumour cells and
99:7054–7059. peripheral nerves studied by in situ hybridization. J
464 Reddy, S.M, R.L Witter, and I. Gimeno. 2000. Gen Virol. 57:285–296.
Development of a quantitative‐competitive 476 Ross, N., M.M Binns, M.J Sanderson, and K.A Schat.
polymerase chain reaction assay for serotype 1 1993. Alterations in DNA sequence and RNA
Marek’s disease virus. Avian Dis. 44:770–775. transcription of the Bam HI‐H fragment accompany
Chapter 15  Neoplastic Diseases 677

attenuation of oncogenic Marek’s disease herpesvirus. disease virus on evaluation of genetic resistance. Poult
Virus Genes. 7:33–51. Sci. 60:2559–2566.
477 Ross, N., G. O’Sullivan, C. Rothwell, G. Smith, S.C 494 Schat, K.A, B.W Calnek, J. Fabricant, and D.L
Burgess, M. Rennie, L.F Lee, and T.F Davison. 1997. Graham. 1985. Pathogenesis of infection with
Marek’s disease virus EcoRI‐Q gene (meq) and a small attenuated Marek’s disease virus strains. Avian Pathol.
RNA antisense to ICP4 are abundantly expressed in 14:127–146.
CD4+ cells and cells carrying a novel lymphoid 495 Schat, K.A, C.‐L.H Chen, B.W Calnek, and D. Char.
marker, AV37, in Marek’s disease lymphomas. J Gen 1991. Transformation of T‐lymphocyte subsets by
Virol. 78 2191–2198. Marek’s disease herpesvirus. J Virol. 65:1408–1413.
478 Ross, N.L 1999. T‐cell transformation by Marek’s 496 Schat, K.A, and H.N Erb. 2014. Lack of evidence that
disease virus. Trends Microbiol. 7:22–29. avian oncogenic viruses are infectious for humans: a
479 Santin, E.R, C.E Shamblin, J.T Prigge, V. review. Avian Dis. 58:345–358.
Arumugaswami, R.L Dienglewicz, and M.S Parcells. 497 Schat, K.A, and C.J Markowski‐Grimsrud. 2001.
2006. Examination of the effect of a naturally Immune responses to Marek’s disease virus infection.
occurring mutation in glycoprotein L on Marek’s Curr Top Microbiol Immunol. 255:91–120.
disease virus pathogenesis. Avian Dis. 50:96–103. 498 Schat, K.A, and V. Nair. 2008. Marek’s disease. In:
480 Sarson, A.J, M.F Abdul‐Careem, L.R Read, J.T Brisbin, Diseases of Poultry, 12th Ed. Y.M Saif, A.M Fadly, J.R
and S. Sharif. 2008. Expression of cytotoxicity‐ Glisson, L.R McDougald, L.K Nolan and D.E Swayne,
associated genes in Marek’s disease virus‐infected eds. Academic Press, Ames. 452–514.
chickens. Viral Immunol. 21:267–272. 499 Schat, K.A, and V. Nair. 2013. Neoplastic diseases:
481 Sarson, A.J, P. Parvizi, D. Lepp, M. Quinton, and S. Marek’s disease. In: Diseases of Poultry, 13th ed. D.E
Sharif. 2008. Transcriptional analysis of host Swayne, J.R Glisson, L.R McDougald, L.K Nolan, D.L
responses to Marek’s disease virus infection in Suarez and V.L Nair, eds. Wyley‐Blackwell, Hoboken,
genetically resistant and susceptible chickens. Animal NJ. 515–552.
Gen. 39:232–240. 500 Schat, K.A, and M.A Skinner. 2008. Avian
482 Schat, K. A Unpublished data. immunosuppressive diseases and immune evasion. In:
483 Schat, K.A 1980. Role of the spleen in the pathogenesis Avian Immunology. F. Davison, B. Kaspers and K.A
of Marek’s disease. Avian Pathol. 10:171–182. Schat, eds. AP Academic Press, London. 299–322.
484 Schat, K.A 1985. Characteristics of the virus. In: 501 Schat, K.A, and V.L van Santen. 2008. Chicken
Marek’s disease. L.N Payne, ed. Martinus Nijhoff, infectious anemia. In: Diseases of Poultry, 13th ed. D.E
Boston. 77–112. Swayne and et al, eds. Wiley‐Blackwell, Ames, IA.
485 Schat, K.A 1997. Prevention of Marek’s disease. World 502 Schat, K.A, and Z. Xing. 2000. Specific and
Poult.:S15–17. nonspecific immune responses to Marek’s disease
486 Schat, K.A 2004. Marek’s disease immunosuppression. virus. Dev Comp Immunol. 24:201–221.
In: Marek’s Disease: An Evolving Problem. F. Davison 503 Schermuly, J., A. Greco, S. Härtle, N. Osterrieder, B.B
and V. Nair, eds. Academic Press, London. 142–155. Kaufer, and B. Kaspers. 2015. In vitro model for lytic
487 Schat, K.A 2005. Isolation of Marek’s disease virus: replication, latency, and transformation of an
revisited. Avian Pathol. 34:91–95. oncogenic alphaherpesvirus. Proc Natl Acad Sci.
488 Schat, K.A 2016. History of the first‐generation 112:7279–7284.
Marek’s disease vaccines: the science and little‐known 504 Schierman, L.W, and O.J Fletcher. 1980. Genetic
facts. Avian Dis. 60:715–724. control of Marek’s disease virus‐induced transient
489 Schat, K.A, and E. Baranowski. 2007. Animal paralysis: association with the major
vaccination and the evolution of viral pathogens. Rev histocompatibility complex. In: Resistance and
Sci Tech Off Int Epiz. 26:327–338. Immunity to Marek’s Disease. P.M Biggs, ed.
490 Schat, K.A, and B.W Calnek. 1978. Characterization Commission European Communities, Luxembourg.
of an apparently nononcogenic Marek’s disease virus. 429–442.
J Natl Cancer Inst. 60:1075–1082. 505 Schmahl, W., G. Hoffmann‐Fezer, and R. Hoffmann.
491 Schat, K.A, B.W Calnek, and J. Fabricant. 1981. 1975. Pathogenesis of neural lesions in Marek’s
Influence of the bursa of Fabricius on the pathogenesis disease. I. Allergic skin reaction against myelin of the
of Marek’s disease. Infect Immun. 31:199–207. peripheral nerves (author’s transl). Z
492 Schat, K.A, B.W Calnek, and J. Fabricant. 1982. Immunitatsforsch Exp Klin Immunol. 150:175–183.
Characterisation of two highly oncogenic strains of 506 Schumacher, D., B.K Tischer, W. Fuchs, and N.
Marek’s disease virus. Avian Pathol. 11:593–605. Osterrieder. 2000. Reconstitution of Marek’s disease
493 Schat, K.A, B.W Calnek, J. Fabricant, and H. virus serotype 1 (MDV‐1) from DNA cloned as a
Abplanalp. 1981. Influence of oncogenicity of Marek’ bacterial artificial chromosome and characterization
678 Section II  Viral Diseases

of a glycoprotein B‐negative MDV‐1 mutant. J Virol. turkeys as embryos or a hatch. Am J Vet Res.
74:11088–11098. 45:1619–1623.
507 Schumacher, D., B.K Tischer, S.M Reddy, and N. 518 Sharma, J.M, L.F Lee, and R.L Witter. 1980. Effect of
Osterrieder. Glycoproteins E and 2001. I of Marek’s neonatal thymectomy on pathogenesis of herpesvirus
disease virus serotype 1 are essential for virus growth of turkeys in chickens. Am J Vet Res. 40:761–764.
in cultured cells. J Virol. 75:11307–11318. 519 Sharma, J.M, R.L Witter, and B.R Burmester. 1973.
508 Schumacher, D., B.K Tischer, J.P Teifke, K. Wink, and Pathogenesis of Marek’s disease in old chickens: lesion
N. Osterrieder. 2002. Generation of a permanent cell regression as the basis for age‐related resistance. Infect
line that supports efficient growth of Marek’s disease Immun. 8:715–724.
virus (MDV) by constitutive expression of MDV 520 Sharma, J.M, R.L Witter, and H.G Purchase. 1975.
glycoprotein E. J Gen Virol. 83:1987–1992. Absence of age‐resistance in neonatally
509 Schumacher, D., B.K Tischer, S. Trapp, and N. thymectomised chickens as evidence for cell‐mediated
Osterrieder. 2005. The protein encoded by the US3 immune surveillance in Marek’s disease. Nature.
orthologue of Marek’s disease virus is required for 253:477–479.
efficient de‐envelopment of perinuclear virions and 521 Shek, W.R, B.W Calnek, K.A Schat, and C.‐L.H Chen.
involved in actin stress fiber breakdown. J Virol. 1983. Characterization of Marek’s disease virus‐
79:3987–3997. infected lymphocytes: Discrimination between
510 Seimon, T.A, D. McAloose, B. Raphael, K.S cytolytically and latently infected cells. J Natl Cancer
Honkavuori, T. Chang, D.L Hirschberg, and W.I Inst. 70:485–491.
Lipkin. 2011. A Novel Herpesvirus in 3 Species of 522 Shigekane, H., Y. Kawaguchi, M. Shirakata, M.
pheasants: Mountain Peacock Pheasant (Polyplectron Sakaguchi, and K. Hirai. 1999. The bi‐directional
inopinatum), Malayan Peacock Pheasant transcriptional promoters for the latency‐relating
(Polyplectron malacense), and Congo Peafowl transcripts of the pp38/pp24 mRNAs and the 1.8 kb‐
(Afropavo congensis). Vet Pathol. Online. mRNA in the long inverted repeats of Marek’s disease
511 Shack, L.A, J.J Buza, and S.C Burgess. 2008. The virus serotype 1 DNA are regulated by common
neoplastically transformed (CD30hi) Marek’s disease promoter‐specific enhancers. Arch Virol.
lymphoma cell phenotype most closely resembles T‐ 144:1893–1907.
regulatory cells. Cancer Immunol Immunother. 523 Shkreli, M., G. Dambrine, D. Soubieux, E. Kut, and D.
57:1253–1262. Rasschaert. 2007. Involvement of the oncoprotein c‐
512 Shamblin, C.E, N. Greene, V. Arumugaswami, R.L Myc in viral telomerase RNA gene regulation during
Dienglewicz, and M.S Parcells. 2004. Comparative Marek’s disease virus‐induced lymphomagenesis.
analysis of Marek’s disease virus (MDV) glycoprotein‐, J Virol. 81:4848–4857.
lytic antigen pp38‐ and transformation antigen Meq‐ 524 Siccardi, F.J, and B.R Burmester. 1970. The differential
encoding genes: association of meq mutations with diagnosis of lymphoid leukosis and Marek’s disease.
MDVs of high virulence. Vet Microbiol. 102:147–167. USDA Tech Bull 1412, Washington, DC.
513 Sharma, J.M 1980. In vitro suppression of T‐cell 525 Silva, R.F 1992. Differentiation of pathogenic and
mitogenic response and tumor cell proliferation by non‐pathogenic serotype 1 Marek’s disease viruses
spleen macrophages from normal chickens. Infect (MDVs) by the polymerase chain reaction
Immun. 28:914–922. amplification of the tandem direct repeats within the
514 Sharma, J.M 1981. Natural killer cell activity in MDV genome. Avian Dis. 36:521–528.
chickens exposed to Marek’s disease virus: inhibition 526 Silva, R.F, J.R Dunn, H.H Cheng, and M. Niikura.
of activity in susceptible chickens and enhancement of 2010. A MEQ‐deleted Marek’s disease virus cloned as
activity in resistant and vaccinated chickens. Avian a bacterial artificial chromosome is a highly
Dis. 25:882–893. efficacious vaccine. Avian Dis. 54:862–869.
515 Sharma, J.M 2008. The avian immune system. In: 527 Silva, R.F, and I.M Gimeno. Oncogenic 2007. Marek’s
Diseases of Poultry, 12th ed. Y.M Saif, A.M Fadly, J.R disease viruses lacking the 132 base pair repeats can
Glisson, L.R McDougald, L.K Nolan and D.E Swayne, still be attenuated by serial in vitro cell culture
eds. Blackwell Publishing, Ames, IA. 47–58. passages. Virus Genes. 34:87–90.
516 Sharma, J.M, and B.R Burmester. 1982. Resistance to 528 Silva, R.F, and R.L Witter. 1985. Genomic expansion
Marek’s disease at hatching in chickens vaccinated as of Marek’s disease virus DNA is associated with serial
embryos with the turkey herpesvirus. Avian Dis. in vitro passage. J Virol. 54:690–696.
26:134–149. 529 Singh, S.M, S.J Baigent, L.J Petherbridge, L.P Smith,
517 Sharma, J.M, L.F Lee, and P.S Wakenell. 1984. and V.K Nair. 2010. Comparative efficacy of BAC‐
Comparative viral, immunologic, and pathologic derived recombinant SB‐1 vaccine and the parent wild
responses of chickens inoculated with herpesvirus of type strain in preventing replication, shedding and
Chapter 15  Neoplastic Diseases 679

disease induced by virulent Marek’s disease virus. Res Marek’s disease: influence of strain of chickens,
Vet Sci. 89:140–145. maternal antibody, and type of vaccine. Avian Dis.
530 Smith, G.D, V. Zelnik, and L.J N. Ross. 1995. Gene 18:33–44.
organization in herpesvirus of turkeys: Identification 543 Spencer, J.L, F. Gilka, J.S Gavora, R.J Hampson, and
of a novel open reading frame in the long unique D.J Caldwell. 1992. Studies with a Marek’s disease
region and a truncated homolog of pp38 in the virus that caused blindness and high mortality in
internal repeat. Virology. 207:205–216. vaccinated flocks. In: Proc 4th Int Symp Marek’s Dis.
531 Smith, J., J.‐R. Sadeyen, I.R Paton, P.M Hocking, N. G. de Boer and S.H M. Jeurissen, eds. Ponsen &
Salmon, M. Fife, V. Nair, D.W Burt, and P. Kaiser. Looijen, Wageningen, Amsterdam, The Netherlands.
2011. Systems analysis of immune responses in 199–201.
Marek’s disease virus‐infected chickens identifies a 544 Stephens, E.A, R.L Witter, K. Nazerian, and J.M
gene involved in susceptibility and highlights a Sharma. 1980. Development and characterization of a
possible novel pathogenicity mechanism. J Virol.. Marek’s disease transplantable tumor in inbred line
85:11146–11158. 72 chickens homozygous at the major (B)
532 Smith, M.W, and B.W Calnek. 1974. Comparative histocompatibility locus. Avian Dis. 24:358–374.
features of low‐virulence and high‐virulence Marek’s 545 Su, S., N. Cui, Y. Zhou, Z. Chen, Y. Li, J. Ding, Y.
disease virus infections. Avian Pathol. 3:229–246. Wang, L. Duan, and Z. Cui. 2015. A recombinant field
533 Smith, M.W, and B.W Calnek. 1974. High virulence strain of Marek’s disease (MD) virus with
Marek’s disease virus infection in chickens previously reticuloendotheliosis virus long terminal repeat insert
infected with low‐virulence virus. J Natl Cancer Inst. lacking the meq gene as a vaccine against MD.
52:1595–1603. Vaccine. 33:596–603.
534 Smith, T.W, D.M Albert, N. Robinson, B.W Calnek, 546 Suchodolski, P.F, Y. Izumiya, B. Lupiani, D.K
and O. Schwabe. 1974. Ocular manifestations of Ajithdoss, O. Gilad, L.F Lee, H.J Kung, and S.M
Marek’s disease. Invest Ophthalmol. 13:586–592. Reddy. 2009. Homodimerization of Marek’s disease
535 Solomon, J.J, and R.L Witter. 1973. Absence of virus‐encoded Meq protein is not sufficient for
Marek’s disease in chicks hatched from eggs transformation of lymphocytes in chickens. J Virol..
containing blood or meat spots. Avian Dis. 83:859–869.
17:141–144. 547 Suchodolski, P.F, Y. Izumiya, B. Lupiani, D.K
536 Solomon, J.J, R.L Witter, H.A Stone, and L.R Ajithdoss, L.F Lee, H.J Kung, and S.M Reddy. 2010.
Champion. 1970. Evidence against embryo Both homo and heterodimers of Marek’s disease virus
transmission of Marek’s disease virus. Avian Dis. encoded Meq protein contribute to transformation of
14:752–762. lymphocytes in chickens. Virology. 399:312–321.
537 Spatz, S.J 2010. Accumulation of attenuating 548 Swayne, D.E, O.J Fletcher, and L.W Schierman. 1988.
mutations in varying proportions within a high Marek’s disease virus‐induced transient paralysis in
passage very virulent plus strain of Gallid herpesvirus chickens: alterations in brain density. Acta
type 2. Virus Res. 149:135–142. Neuropathol. 76:287–291.
538 Spatz, S.J, L. Petherbridge, Y. Zhao, and V. Nair. 2007. 549 Swayne, D.E, O.J Fletcher, and L.W Schierman. 1989.
Comparative full‐length sequence analysis of Marek’s disease virus‐induced transient paralysis in
oncogenic and vaccine (Rispens) strains of Marek’s chickens. 1. Time course association between clinical
disease virus. J Gen Virol. 88:1080–1096. signs and histological brain lesions. Avian Pathol.
539 Spatz, S.J, C. Rue, D. Schumacher, and N. Osterrieder. 18:385–396.
2008. Clustering of mutations within the inverted 550 Swayne, D.E, O.J Fletcher, and L.W Schierman. 1989.
repeat regions of a serially passaged attenuated gallid Marek’s disease virus‐induced transient paralysis in
herpesvirus type 2 strain. Virus Genes. 37:69–80. chickens: Demonstration of vasogenic brain oedema
540 Spatz, S.J, and K.A Schat. 2011. Comparative genomic by an immunohistochemical method. J Comp Path.
sequence analysis of the Marek’s disease vaccine strain 101:451–462.
SB‐1. Virus Genes. 42:331–338. 551 Tai, S.S, C. Hearn, S. Umthong, O. Agafitei, H.H
541 Spatz, S.J, L.P Smith, S.J Baigent, L. Petherbridge, and Cheng, J.R Dunn, and M. Niikura. 2017. Expression
V. Nair. 2011. Genotypic characterization of two of Marek’s disease virus oncoprotein Meq during
bacterial artificial chromosome clones derived from a infection in the natural host. Virology.
single DNA source of the very virulent gallid 503:103–113.
herpesvirus‐2 strain C12/130. J Gen Virol. 552 Tan, X., P. Brunovskis, and L.F Velicer. 2001.
92:1500–1507. Transcriptional analysis of Marek’s disease virus
542 Spencer, J.L, J.S Gavora, A.A Grunder, A. Robertson, glycoprotein D, I, and E genes: gD expression is
and G.W Speckmann. 1974. Immunization against undetectable in cell culture. J Virol. 75:2067–2075.
680 Section II  Viral Diseases

553 Tanaka, A., S. Silver, and M. Nonoyama. 1978. 565 Trapp, S., M.S Parcells, J.P Kamil, D. Schumacher, B.K
Biochemical evidence of the nonintegrated status of Tischer, P.M Kumar, V.K Nair, and N. Osterrieder.
Marek’s disease virus DNA in virus‐transformed 2006. A virus‐encoded telomerase RNA promotes
lymphoblastoid cells of chickens. Virology. 88:19–24. malignant T cell lymphomagenesis. J Exp Med.
554 Tarpey, I., P.J Davis, P. Sondermeijer, C. van Geffen, I. 203:1307–1317.
Verstegen, V.E Schijns, J. Kolodsick, and R. Sundick. 566 Tsukamoto, K., C. Kojima, Y. Komori, N. Tanimura,
2007. Expression of chicken interleukin‐2 by turkey M. Mase, and S. Yamaguchi. 1999. Protection of
herpesvirus increases the immune response against chickens against very virulent infectious bursal disease
Marek’s disease virus but fails to increase protection virus (IBDV) and Marek’s disease virus (MDV) with a
against virulent challenge. Avian Pathol. 36:69–74. recombinant MDV expressing IBDV VP2. Virology.
555 Tavlarides‐Hontz, P., P.M Kumar, J.R Amortegui, N. 257:352–362.
Osterrieder, and M.S Parcells. 2009. A deletion within 567 Tulman, E.R, C.L Afonso, Z. Lu, L. Zsak, D.L Rock,
glycoprotein L of Marek’s disease virus (MDV) field and G.F Kutish. 2000. The genome of a very virulent
isolates correlates with a decrease in bivalent MDV Marek’s disease virus. J Virol. 74:7980–7988.
vaccine efficacy in contact‐exposed chickens. Avian 568 Vagnozzi, A., G. Zavala, S.M Riblet, A. Mundt, and
Dis. 53:287–296. M. Garcia. 2012. Protection induced by
556 Teng, M., Z.H Yu, P. Zhao, G.Q Zhuang, Z.X Wu, L. commercially available live‐attenuated and
Dang, H.Z Li, S.M Ma, Z.Z Cui, G.P Zhang, R. Wu, recombinant viral vector vaccines against infectious
and J. Luo. 2017. Putative roles as oncogene or tumour laryngotracheitis virus in broiler chickens. Avian
suppressor of the Mid‐clustered microRNAs in Gallid Pathol. 41:21–31.
alphaherpesvirus 2 (GaHV2) induced Marek’s disease 569 Vallejo, R.L, L.D Bacon, H.C Liu, R.L Witter, M.A
lymphomagenesis. J Gen Virol. 98:1097–1112. Groenen, J. Hillel, and H.H Cheng. 1998. Genetic
557 Thanthrige‐Don, N., L.R Read, M.F Abdul‐Careem, H. mapping of quantitative trait loci affecting
Mohammadi, A.I Mallick, and S. Sharif. 2010. Marek’s susceptibility to Marek’s disease virus induced tumors
disease virus influences the expression of genes in F2 intercross chickens. Genetics. 148:349–360.
associated with IFN‐gamma‐inducible MHC class II 570 Veiga, I.B, K.W Jarosinski, B.B Kaufer, and N.
expression. Viral Immunol. 23:227–232. Osterrieder. 2013. Marek’s disease virus (MDV)
558 Theis, G.A, L.W Schierman, and R.A McBride. 1974. ubiquitin‐specific protease (USP) performs critical
Transplantation of a Marek’s disease lymphoma in functions beyond its enzymatic activity during virus
syngeneic chickens. J Immunol. 113:1710–1715. replication. Virology. 437:110–117.
559 Thornton, D.H 1985. Quality control and 571 Venugopal, K., A.P Bland, L.J N. Ross, and L.N Payne.
standardization of vaccines. In: Marek’s Disease. L.N 1996. Pathogenicity of an unusual highly virulent
Payne, ed. Martinus Nijhoff, Boston, MA. 267–291. Marek’s disease virus isolated in the United Kingdom.
560 Thurston, T.J, R.A Hess, H.K Adldinger, R.F In: Current Research on Marek’s Disease. R.F Silva,
Solorzano, and H.V Biellier. 1975. Ultrastructural H.H Cheng, P.M Coussens, L.F Lee and L.F Velicer,
studies of semen abnormalities and herpesvirus eds. American Association of Avian Pathologists,
associated with cultured testis cells from domestic Kennett Square, PA. 119–124.
turkeys. J Reprod Fertil. 45:507–514. 572 Vielitz, E., and H. Landgraf. 1986. Protection against
561 Tian, F., J. Luo, H. Zhang, S. Chang, and J. Song. 2012. Marek’s disease with different vaccines, determination
MiRNA expression signatures induced by Marek’s of PD50 and duration of vaccinal immunity. Dtsch
disease virus infection in chickens. Genomics. Tierarztl Wochenschr. 93:53–55.
99:152–159. 573 Volpini, L.M, B.W Calnek, M.J Sekellick, and P.I
562 Tischer, B.K, and N. Osterrieder. Herpesviruses—2010. Marcus. 1995. Stages of Marek’s disease virus latency
A zoonotic threat? Vet Microbiol. 140:266–270. defined by variable sensitivity to interferon
563 Tischer, B.K, D. Schumacher, M. Beer, J. Beyer, J.P modulation of viral antigen expression. Vet Microbiol.
Teifke, K. Osterrieder, K. Wink, V. Zelnik, F. Fehler, 47:99–109.
and N. Osterrieder. 2002. A DNA vaccine containing 574 von Bülow, V. 1971. Diagnosis and certain biological
an infectious Marek’s disease virus genome can confer properties of the virus of Marek’s disease. Am J Vet
protection against tumorigenic Marek’s disease in Res. 32:1275–1288.
chickens. J Gen Virol. 83:2367–2376. 575 von Bülow, V. 1977. Further characterisation of the
564 Tischer, B.K, D. Schumacher, M. Messerle, M. Wagner, CVI 988 strain of Marek’s disease virus. Avian Pathol.
and N. Osterrieder. 2002. The products of the UL10 6:395–403.
(gM) and the UL49.5 genes of Marek’s disease virus 576 von Bülow, V., and P.M Biggs. 1975. Precipitating
serotype 1 are essential for virus growth in cultured antigens associated with Marek’s disease viruses and a
cells. J Gen Virol. 83:997–1003. herpesvirus of turkeys. Avian Pathol. 4:147–162.
Chapter 15  Neoplastic Diseases 681

577 von Bülow, V., and P.M Biggs. 1975. Differentiation 589 Witter, R.L 1982. Protection by attenuated and
between strains of Marek’s disease virus and turkey polyvalent vaccines against highly virulent strains of
herpesvirus by immunofluorescence assays. Avian Marek’s Disease virus. Avian Pathol. 11:49–62.
Pathol. 4:133–146. 590 Witter, R.L 1983. Characteristics of Marek’s disease
578 von Bülow, V., R. Rudolph, and B. Fuchs. Erhöhte viruses isolated from vaccinated commercial chicken
1986. Pathogenität des Erregers der aviären flocks: association of viral pathotype with lymphoma
infektiösen Anämia bei Hühnerküken (CAA) bei frequency. Avian Dis. 27:113–132.
simultaner Infektion mit Virus der Marekschen 591 Witter, R.L 1987. New serotype 2 and attenuated
Krankheit (MDV), Bursitisvirus (IBDV) oder serotype 1 Marek’s disease vaccine viruses:
Reticuloendotheliosevirus (REV). Zentralbl comparative efficacy. Avian Dis. 31:752–765.
Veterinarmed. 33:93–116. 592 Witter, R.L 1989. Very virulent Marek’s disease
579 Waidner, L.A, R.W Morgan, A.S Anderson, E.L viruses: importance and control. World’s Poult Sci. J
Bernberg, S. Kamboj, M.T Garcia, S.M Riblet, M. 45:60–65.
Ouyang, G.K Isaacs, M. Markis, B.C Meyers, P.J 593 Witter, R.L 1992. Influence of serotype and virus
Green, and J. Burnside. 2009. MicroRNAs of Gallid strain on synergism between Marek’s disease vaccine
and Meleagrid herpesviruses show generally viruses. Avian Pathol. 21:601–614.
conserved genomic locations and are virus‐specific. 594 Witter, R.L 1995. Attenuation of lymphoid leukosis
Virology. 388:128–136. enhancement by serotype 2 Marek’s disease virus.
580 Wakenell, P.S, T. Bryan, J. Schaeffer, A. Avakian, C. Avian Pathol. 24:665–678.
Williams, and C. Whitfill. 2002. Effect of in ovo 595 Witter, R. 1995. L. personal communication.
vaccine delivery route on herpesvirus of turkeys/SB‐1 596 Witter, R.L 1996. Historic incidence of Marek’s disease
efficacy and viremia. Avian Dis. 46:274–280. as revealed by condemnation statistics. In: 5th
581 Walkden‐Brown, S.W, A. Islam, P.J Groves, A. International Symposium on Marek’s Disease. East
Rubite, S.M Sharpe, and S.K Burgess. 2013. Lansing, MI. 501–508.
Development, application, and results of routine 597 Witter, R.L 1997. Increased virulence of Marek’s
monitoring of Marek’s disease virus in broiler house disease virus field isolates. Avian Dis. 41:149–163.
dust using real‐time quantitative PCR. Avian Dis. 598 Witter, R.L 1998. The changing landscape of Marek’s
57:544–554. disease. Avian Pathol. 27:S46–S53.
582 Walkden‐Brown, S.W, A. Islam, A.F Islam, S.K 599 Witter, R.L 2001. Marek’s disease vaccines ‐ past,
Burgess, P.J Groves, and J. Cooke. 2013. Pathotyping present and future (chicken vs virus—a battle of the
of Australian isolates of Marek’s disease virus in centuries). In: Current Progress on Marek’s Disease
commercial broiler chickens vaccinated with Research. K.A Schat, R.W Morgan, M.S Parcells and
herpesvirus of turkeys (HVT) or bivalent (HVT/SB1) J.L Spencer, eds. American Association of Avian
vaccine and association with viral load in the spleen Pathologists, Kennett Square, PA. 1–9.
and feather dander. Aust Vet J. 91:341–350. 600 Witter, R.L 2001. Protective efficacy of Marek’s
583 Wang, X., X. Chi, and M. Wang. 2011. Structural disease vaccines. Curr Top Microbiol Immunol.
characteristics and antiviral activity of multiple 255:57–90.
peptides derived from MDV glycoproteins B and H. 601 Witter, R.L 2005. Marek’s disease: the continuing
Virol J. 8:190. struggle between pathogen and host. Vet J.
584 Weli, S.C, and M. Tryland. 2011. Avipoxviruses: 170:149–150.
infection biology and their use as vaccine vectors. 602 Witter, R.L, G.H Burgoyne, and B.R Burmester. 1968.
Virol J. 8:49. Survival of Marek’s disease agent in litter and
585 Williams, C.J, and B.A Hopkins. 2011. Field evaluation droppings. Avian Dis. 12:522–530.
of the accuracy of vaccine deposition by two different 603 Witter, R.L, and B.R Burmester. 1979. Differential
commercially available in ovo injection systems. Poult effect of maternal antibodies on efficacy of cellular
Sci. 90:223–226. and cell‐free Marek’s disease vaccines. Avian Pathol.
586 Williams, C.J, and A.S Zedek. 2010. Comparative field 8:145–156.
evaluations of in ovo applied technology. Poult Sci. 604 Witter, R.L, B.W Calnek, C. Buscaglia, I.M Gimeno,
89:189–193. and K.A Schat. 2005. Classification of Marek’s disease
587 Wilson, M.R, and P.M Coussens. 1991. Purification viruses according to pathotype: philosophy and
and characterization of infectious Marek’s disease methodology. Avian Pathol. 34:75–90.
virus genomes using pulsed field electrophoresis. 605 Witter, R.L, and I. Gimeno. 2006. Susceptibility of
Virology. 185:673–680. adult chickens, with and without prior vaccination, to
588 Witter, R.L 1972. Turkey herpesvirus: lack of challenge with Marek’s disease virus. Avian Dis.
oncogenicity for turkeys. Avian Dis. 16:666–670. 50:354–365.
682 Section II  Viral Diseases

606 Witter, R.L, I.M Gimeno, A.R Pandiri, and A.M Fadly. 619 Witter, R.L, and J.J Solomon. 1971. Epidemiology of a
2010. Tumor diagnosis manual: The differential herpesvirus of turkeys: Possible sources and spread
diagnosis of lymphoid and myeloid tumors in the of infection in turkey flocks. Infect Immun
chicken. American Association of Avian Pathologists, 4:356–361.
Jacksonville, FL. 620 Witter, R.L, and J.J Solomon. 1972. Experimental
607 Witter, R.L, I.M Gimeno, W.M Reed, and L.D Bacon. infection of turkeys and chickens with a herpesvirus
1999. An acute form of transient paralysis induced by of turkeys (HVT). Avian Dis. 16:34–44.
highly virulent strains of Marek’s disease virus. Avian 621 Witter, R.L, J.J Solomon, and G.H Burgoyne. 1969.
Dis. 43:704–720. Cell culture techniques for primary isolation of
608 Witter, R.L, and K.S Kreager. 2004. Serotype 1 viruses Marek’s disease‐associated herpesvirus. Avian Dis.
modified by backpassage or insertional mutagenesis: 13:101–118.
Approaching the threshold of vaccine efficacy in 622 Witter, R.L, J.J Solomon, L.R Champion, and K.
Marek’s disease. Avian Dis. 48:768–782. Nazerian. 1971. Long term studies of Marek’s disease
609 Witter, R.L, and L.F Lee. Polyvalent 1984. Marek’s infection in individual chickens. Avian Dis.
disease vaccines: Safety, efficacy and protective 15:346–365.
synergism in chickens with maternal antibodies. 623 Witter, R.L, E.A Stephens, J.M Sharma, and K.
Avian Pathol. 13:75–92. Nazerian. 1975. Demonstration of a tumor‐associated
610 Witter, R.L, L.F Lee, L.D Bacon, and E.J Smith. 1979. surface antigen in Marek’s disease. J Immunol.
Depression of vaccinal immunity to Marek’s disease 115:177–183.
by infection with reticuloendotheliosis virus. Infect 624 Wolc, A., J. Arango, T. Jankowski, P. Settar, J.E Fulton,
Immun. 26:90–98. N.P O’Sullivan, R. Fernando, D.J Garrick, and J.C
611 Witter, R.L, D. Li, D. Jones, L.F Lee, and H.J Kung. Dekkers. 2013. Genome‐wide association study for
1997. Retroviral insertional mutagenesis of a Marek’s disease mortality in layer chickens. Avian Dis.
herpesvirus: a Marek’s disease virus mutant 57:395–400.
attenuated for oncogenicity but not for 625 Wozniakowski, G., and J.S Niczyporuk. 2015.
immunosuppression or in vivo replication. Avian Dis. Detection of specific UL49 sequences of Marek’s
41:407–421. disease virus CVI988/Rispens strain using loop‐
612 Witter, R.L, J.I Moulthrop Jr., G.H Burgoyne, and H.C mediated isothermal amplification. J Virol Methods.
Connell. 1970. Studies on the epidemiology of Marek’s 221:22–28.
disease herpesvirus in broiler flocks. Avian Dis. 626 Wozniakowski, G., and A.E Samorek‐Salamonowicz.
14:255–267. 2014. Molecular evolution of Marek’s disease virus
613 Witter, R.L, K. Nazerian, H.G Purchase, and G.H (MDV) field strains in a 40‐year time period. Avian
Burgoyne. 1970. Isolation from turkeys of a cell‐ Dis. 58:550–557.
associated herpesvirus antigenically related to Marek’s 627 Wozniakowski, G., E. Samorek‐Salamonowicz, and W.
disease virus. Am J Vet Res. 31:525–538. Kozdrun. 2013. Comparison of loop‐mediated
614 Witter, R.L, and L. Offenbecker. 1979. Nonprotective isothermal amplification and PCR for the detection
and temperature‐sensitive variants of Marek’s disease and differentiation of Marek’s disease virus serotypes
vaccine viruses. J Natl Cancer Inst. 62:143–151. 1, 2, and 3. Avian Dis. 57:539–543.
615 Witter, R.L, J.M Sharma, and A.M Fadly. 1980. 628 Woźniakowski, G.J, E. Samorek‐Salamonowicz, and
Pathogenicity of variant Marek’s disease virus isolants W. Kozdruń. 2011. Rapid detection of Marek’s disease
in vaccinated and unvaccinated chickens. Avian Dis. virus in feather follicles by loop‐mediated
24:210–232. amplification (LAMP). Avian Dis. 55:462–467.
616 Witter, R.L, J.M Sharma, L.F Lee, H.M Opitz, and 629 Wu, C., J. Gan, Q. Jin, C. Chen, P. Liang, Y. Wu, X. Liu,
C.W Henry. 1984. Field trials to test the efficacy of L. Ma, and F. Davison. 2009. Revaccination with
polyvalent Marek’s disease vaccines in broilers. Avian Marek’s disease vaccines induces productive infection
Dis. 28:44–60. and superior immunity. Clin Vaccine Immunol.
617 Witter, R.L, J.M Sharma, and L. Offenbecker. 1976. 16:184–193.
Turkey herpesvirus infection in chickens: Induction of 630 Xing, Z., and K.A Schat. 2000. Expression of cytokine
lymphoproliferative lesions and characterization of genes in Marek’s disease virus‐infected chickens and
vaccinal immunity against Marek’s disease. Avian Dis. chicken embryo fibroblast cultures. Immunol.
20:676–692. 100:70–76.
618 Witter, R.L, R.F Silva, and L.F Lee. 1987. New 631 Xing, Z., and K.A Schat. 2000. Inhibitory effects of
serotype 2 and attenuated serotype 1 Marek’s disease nitric oxide and gamma interferon on in vitro and in
vaccine viruses: selected biological and molecular vivo replication of Marek’s disease virus. J Virol.
characteristics. Avian Dis. 31:829–840. 74:3605–3612.
Chapter 15  Neoplastic Diseases 683

632 Xu, S., N. Yonash, R.L Vallejo, and H.H Cheng. 1998. 645 Zelnik, V. 2003. Marek’s disease virus research in the
Mapping quantitative trait loci for binary traits using post‐sequencing era: new tools for the study of gene
a heterogeneous residual variance model: an functions and virus‐host interactions. Avian Pathol.
application to Marek’s disease susceptibility in 32:323–334.
chickens. Genetica. 104:171–178. 646 Zelnik, V., O. Harlin, F. Fehler, B. Kaspers, T.W Gobel,
633 Yamaguchi, T., S.L Kaplan, P. Wakenell, and K.A V.K Nair, and N. Osterrieder. 2004. An enzyme‐linked
Schat. 2000. Transactivation of latent Marek’s disease immunosorbent assay (ELISA) for detection of
herpesvirus genes in QT35, a quail fibroblast cell line, Marek’s disease virus‐specific antibodies and its
by herpesvirus of turkeys. J Virol. 74:10176–10186. application in an experimental vaccine trial. J Vet Med
634 Yan, Y., N. Yang, H.H Cheng, J. Song, and L. Qu. 2015. B Infect Dis Vet Public Health. 51:61–67.
Genome‐wide identification of copy number 647 Zhang, X., Y. Wu, Y. Huang, and X. Liu. 2012.
variations between two chicken lines that differ in Protection conferred by a recombinant Marek’s
genetic resistance to Marek’s disease. BMC Genomics. disease virus that expresses the spike protein from
16:843. infectious bronchitis virus in specific pathogen‐free
635 Yao, Y., and V. Nair. 2014. Role of virus‐encoded chicken. Virol J. 9:85.
microRNAs in Avian viral diseases. Viruses. 648 Zhang, Y., N. Cui, N. Han, J. Wu, Z. Cui, and S. Su.
6:1379–1394. 2017. Depression of vaccinal immunity to Marek’s
636 Yao, Y., Y. Zhao, L.P Smith, C.H Lawrie, N.J Saunders, disease by infection with chicken infectious anemia
M. Watson, and V. Nair. 2009. Differential expression virus. Front Microbiol. 8:1863.
of microRNAs in Marek’s disease virus‐transformed 649 Zhang, Y., and J.M Sharma. 2001. Early posthatch
T‐lymphoma cell lines. J Gen Virol. 90:1551–1559. protection against Marek’s disease in chickens
637 Yao, Y., Y. Zhao, L.P Smith, M. Watson, and V. Nair. vaccinated in ovo with a CVI988 serotype 1 vaccine.
Novel MicroRNAs (miRNAs) 2009. Encoded by Avian Dis. 45:639–645.
Herpesvirus of Turkeys: evidence of miRNA evolution 650 Zhang, Y., and J.M Sharma. 2003. Immunological
by duplication. J Virol.. 83:6969–6973. tolerance in chickens hatching from eggs injected with
638 Yao, Y., Y. Zhao, H. Xu, L.P Smith, C.H Lawrie, A. cell‐associated herpesvirus of Turkey (HVT). Dev
Sewer, M. Zavolan, and V. Nair. 2007. Marek’s disease Comp Immunol. 27:431–438.
virus type 2 (MDV‐2)‐encoded microRNAs show no 651 Zhang, Z., W. Chen, C. Ma, P. Zhao, L. Duan, F.
sequence conservation to those encoded by MDV‐1. Zhang, A. Sun, Y. Li, H. Su, S. Li, H. Cui, and Z. Cui.
J Virol. 2014. Construction of recombinant Marek’s disease
639 Yao, Y., Y. Zhao, H. Xu, L.P Smith, C.H Lawrie, M. virus (MDV) lacking the meq oncogene and co‐
Watson, and V. Nair. 2008. MicroRNA profile of expressing AIV‐H9N2 HA and NA genes under
Marek’s disease virus‐transformed T‐cell line MSB‐1: control of exogenous promoters. J Biotechnol. 181:45–54.
Predominance of virus‐encoded microRNAs. J Virol.. 652 Zhao, Y., H. Xu, Y. Yao, L.P Smith, L. Kgosana, J.
82:4007–4015. Green, L. Petherbridge, S.J Baigent, and V. Nair. 2011.
640 Yu, Y., J. Luo, A. Mitra, S. Chang, F. Tian, H. Zhang, P. Critical role of the virus‐encoded microRNA‐155
Yuan, H. Zhou, and J. Song. 2011. Temporal ortholog in the induction of Marek’s disease
transcriptome changes induced by MDV in Marek’s lymphomas. PLoS Pathog. 7:e1001305.
disease‐resistant and ‐susceptible inbred chickens. 653 Zhao, Y., Y. Yao, H. Xu, L. Lambeth, L.P Smith, L.
BMC Genomics. 12:501. Kgosana, X. Wang, and V. Nair. 2009. A functional
641 Yuasa, N. 1983. Propagation and infectivity titration of MicroRNA‐155 ortholog encoded by the oncogenic
the GIFU‐1 strain of chicken anemia agent in a cell Marek’s disease virus. J Virol. 83:489–492.
line (MDCC‐MSB1) derived from Marek’s disease 654 Zhu, G.S, A. Iwata, M. Gong, S. Ueda, and K. Hirai.
lymphoma. Natl Inst Anim Hlth Q. 23:13–20. 1994. Marek’s disease virus type 1‐specific
642 Yunis, R., K.W Jarosinski, and K.A Schat. 2004. phosphorylated proteins pp38 and pp24 with
Association between rate of viral genome replication common amino acid termini are encoded from the
and virulence of Marek’s disease herpesvirus strains. opposite junction regions between the long unique
Virology. 328:142–150. and inverted repeat sequences of viral genome.
643 Zander, D.V 1959. Experiences with epidemic tremor Virology. 200:816–820.
control. In: Proc 8th Annual Western Poultry Disease 655 Zhu, G.S, T. Ojima, T. Hironaka, T. Ihara, N.
Conference. Davis, CA. 18–23. Mizukoshi, A. Kato, S. Ueda, and K. Hirai. 1992.
644 Zander, D.V, R.W Hill, R.G Raymond, R.K Balch, R.W Differentiation of oncogenic and nononcogenic strains
Mitchell, and J.W Dunsing. 1972. The use of blood of Marek’s disease virus type 1 by using polymerase
from selected chickens as an immunizing agent for chain reaction DNA amplification. Avian Dis.
Marek’s disease. Avian Dis. 16:163–178. 36:637–645.
684 Section II  Viral Diseases

656 Zhuang, G., A. Sun, M. Teng, and J. Luo. A Tiny. 2017. 11 Bacon, L.D 2000. Detection of endogenous avian
RNA that Packs a big punch: the critical role of a viral leukosis virus envelope in chicken plasma using R2
miR‐155 ortholog in lymphomagenesis in Marek’s antiserum. Avian Pathol. 29:153–164.
disease. Front Microbiol. 8:1169. 12 Bacon, L.D, T.L Fredericksen, D.G Gilmour, A.M Fadly,
and L.B Crittenden. 1985. Tests of association of
lymphocyte alloantigen genotypes with resistance to
Leukosis/Sarcoma Group
viral oncogenesis in chickens. 2. Rous sarcoma and
1 Adams, M.J, E.J Lefkowitz, A.M Q. King, B. Harrach, lymphoid leukosis in progeny derived from 6(3) X 15(1)
R.L Harrison, N.J Knowles, A.M Kropinski, M. and 100 X 6(3) crosses. Poult Sci. 64:39–47.
Krupovic, J.H Kuhn, A.R Mushegian, M. Nibert, S. 13 Bacon, L.D, E. Smith, L.B Crittenden, and G.B
Sabanadzovic, H. Sanfacon, S.G Siddell, P. Simmonds, Havenstein. 1988. Association of the slow feathering
A. Varsani, F.M Zerbini, A.E Gorbalenya, and A.J (K) and an endogenous viral (ev21) gene on the Z
Davison. 2017. Changes to taxonomy and the chromosome of chickens. Poult Sci. 67:191–197.
International Code of Virus Classification and 14 Bacon, L.D, E.J Smith, A.M Fadly, and L.B Crittenden.
Nomenclature ratified by the International Committee 1996. Development of an alloantiserum (R2) that
on Taxonomy of Viruses. (2017). Arch Virol. detects susceptibility of chickens to subgroup E
162:2505–2538. endogenous avian leukosis virus. Avian Pathol.
2 Adkins, H.B, S.C Blacklow, and J.A Young. 2001. Two 25:551–568.
functionally distinct forms of a retroviral receptor 15 Bacon, L.D, R.L Witter, L.B Crittenden, A. Fadly, and J.
explain the nonreciprocal receptor interference among Motta. 1981. B‐haplotype influence on Marek’s disease,
subgroups B, D, and E avian leukosis viruses. J Virol. Rous sarcoma, and lymphoid leukosis virus‐induced
75:3520–3526. tumors in chickens. Poult Sci. 60:1132–1139.
3 Adkins, H.B, J. Brojatsch, J. Naughton, M.M Rolls, J.M 16 Bacon, L.D, R.L Witter, and A.M Fadly. 1989.
Pesola, and J.A Young. 1997. Identification of a cellular Augmentation of retrovirus‐induced lymphoid leukosis
receptor for subgroup E avian leukosis virus. Proc Natl by Marek’s disease herpesviruses in White Leghorn
Acad Sci USA. 94:11617–11622. chickens. J Virol. 63:504–512.
4 Adkins, H.B, J. Brojatsch, and J.A Young. 2000. 17 Bai, J., K. Howes, L.N Payne, and M.A Skinner. 1995.
Identification and characterization of a shared TNFR‐ Sequence of host‐range determinants in the env gene of
related receptor for subgroup B, D, and E avian leukosis a full‐length, infectious proviral clone of exogenous
viruses reveal cysteine residues required specifically for avian leukosis virus HPRS‐103 confirms that it
subgroup E viral entry. J Virol. 74:3572–3578. represents a new subgroup (designated J). J Gen Virol.
5 Arshad, S.S, A.P Bland, S.M Hacker, and L.N Payne. 76 (Pt 1):181–187.
1997. A low incidence of histiocytic sarcomatosis 18 Bai, J., L.N Payne, and M.A Skinner. 1995. HPRS‐103
associated with infection of chickens with the (exogenous avian leukosis virus, subgroup J) has an env
HPRS‐103 strain of subgroup J avian leukosis virus. gene related to those of endogenous elements EAV‐0
Avian Dis. 41:947–956. and E51 and an E element found previously only in
6 Arshad, S.S, K. Howes, G.S Barron, L.M Smith, P.H sarcoma viruses. J Virol. 69:779–784.
Russell, and L.N Payne. 1997. Tissue tropism of the 19 Ballandras, A., K. Moreau, X. Robert, M.P Confort, R.
HPRS‐103 strain of J subgroup avian leukosis virus and Merceron, R. Haser, C. Ronfort, and P. Gouet. 2011. A
of a derivative acutely transforming virus. Vet Pathol. crystal structure of the catalytic core domain of an
34:127–137. avian sarcoma and leukemia virus integrase suggests an
7 Arshad, S.S, L.M Smith, K. Howes, P.H Russell, K. alternate dimeric assembly. PLoS One. 6:e23032.
Venugopal, and L.N Payne. 1999. Tropism of subgroup 20 Baluda, M.A 1962. Properties of cells infected with
J avian leukosis virus as detected by in situ avian myeloblastosis virus. Cold Spring Harb Symp
hybridization. Avian Pathol. 28:163–169. Quant Biol. 27:415–425.
8 Astrin, S.M, E.G Buss, and W.S Haywards. 1979. 21 Baluda, M.A 1963. Conversion of cells by avian
Endogenous viral genes are non‐essential in the myeloblastosis virus. Perspect Virol. 3:118–137.
chicken. Nature. 282:339–341. 22 Baluda, M.A, and P.P Jamieson. 1961. In vivo infectivity
9 Baba, T.W, and E.H Humphries. 1985. Formation of a studies with avian myeloblastosis virus. Virology. I
transformed follicle is necessary but not sufficient for 14:33–45.
development of an avian leukosis virus‐induced 23 Barbosa, T., M. Ramirez, S. Hafner, S. Cheng, and G.
lymphoma. Proc Natl Acad Sci USA. 82:213–216. Zavala. 2010. Forensic investigation of a 1986 outbreak
10 Baba, T.W, and E.H Humphries. 1986. Selective of osteopetrosis in commercial brown layers reveals a
integration of avian leukosis virus in different novel avian leukosis virus‐related genome. Avian Dis.
hematopoietic tissues. Virology. 155:557–566. 54:981–989.
Chapter 15  Neoplastic Diseases 685

24 Barbosa, T., G. Zavala, and S. Cheng. 2008. Molecular 38 Bova, C.A, J.C Olsen, and R. Swanstrom. 1988. The
characterization of three recombinant isolates of avian avian retrovirus env gene family: molecular analysis of
leukosis virus obtained from contaminated Marek’s host range and antigenic variants. J Virol. 62:75–83.
disease vaccines. Avian Dis. 52:245–252. 39 Boyde, A., A.J Banes, R.M Dillaman, and G.L
25 Barnard, R.J, D. Elleder, and J.A Young. 2006. Avian Mechanic. 1978. Morphological study of an avian bone
sarcoma and leukosis virus‐receptor interactions: from disorder caused by myeloblastosis‐associated virus.
classical genetics to novel insights into virus‐cell Metab Bone Dis Relat Res. 1:235–242.
membrane fusion. Virology. 344:25–29. 40 Brandvold, K.A, P. Neiman, and A. Ruddell. 2000.
26 Bauer, H., and B. Fleischer. 1981. Immunobiology of Angiogenesis is an early event in the generation of
avian RNA tumor virus‐induced cell surface antigens. myc‐induced lymphomas. Oncogene. 19:2780–2785.
In J.W Blasecki (ed.). Mechanisms of Immunity to 41 Brojatsch, J., J. Naughton, H.B Adkins, and J.A Young.
Virus‐Induced Tumors. Marcel Dekker: New York. 69–118. 2000. TVB receptors for cytopathic and noncytopathic
27 Bauer, H., R. Kirth, L. Rohrschneider, and H. subgroups of avian leukosis viruses are functional death
Gelderblum. 1976. Immune response to oncornaviruses receptors. J Virol. 74:11490–11494.
and tumor‐associated antigens in the chicken. Cancer 42 Brothwell, D.R 2002. Ancient avian osteopetrosis: the
Res. 36:598–602. current state of knowledge. Acta Zoologica
28 Beard, J.W 1963. Avian virus growths and their Cracoviensia. 45:315–318.
etiological agents. Adv Cancer Res. 7:1–127. 43 Brown, C.Y, S.J Bowers, G. Loring, C. Heberden, R.M
29 Beard, J.W 1980. Biology of avian oncornaviruses. In: Lee, and P.E Neiman. 2004. Role of Mtd/Bok in normal
Viral Oncology. G. Klein, ed. Raven Press, New York. and neoplastic B‐cell development in the bursa of
55–87. Fabricius. Dev Comp Immunol. 28:619–634.
30 Beard, J.W, J.F Chabot, D. Beard, U. Heine, and G.E 44 Brown, T.P, N. Stedman, and M. Pantin‐Vera. 2000.
Houts. 1976. Renal neoplastic response to leukosis Proceedings, International Symposium on ALV‐J and
virus strains BAI A (avian myeloblastosis virus) and Other Avian Retroviruses. Rauischholzhausen,
MC29. Cancer Res. 36:339–353. Germany. In E.F Kaleta, L.N Payne, and U. Heffels‐
31 Beaudreau, G.S, and C. Becker. 1958. Virus of avian Redmann (eds.) 63–66.
myeloblastosis. X. Photometric microdetermination of 45 Burmester, B.R 1947. Studies on the transmission of
adenosinetriphosphatase activity. J Natl Cancer Inst. avian visceral lymphomatosis. II. Propagation of
20:339–349. lymphomatosis with cellular and cell‐free preparations.
32 Benkel, B., and K. Rutherford. 2014. Endogenous avian Cancer Res. 7:786–797.
leukosis viral loci in the Red Jungle Fowl genome 46 Burmester, B.R 1955. Immunity to visceral
assembly. Poult Sci. 93:2988–2990. lymphomatosis in chicks following injection of virus
33 Bennett, D.D, and S.E Wright. 1987. Immunization into dams. Proc Soc Exp Biol Med. 88:153–155.
with envelope glycoprotein of an avian RNA tumor 47 Burmester, B.R 1968. Unpublished data.
virus protects against sarcoma virus tumor induction: 48 Burmester, B.R, A.K Fontes, and W.G Walter. 1960.
role of subgroup. Virus Res. 8:73–77. Pathogenicity of a viral strain (RPL 12) causing avian
34 Biggs, P.M, and L.N Payne. 1964. Relationship of visceral lymphomatosis and related neoplasms III.
Marek’s disease (neural lymphomatosis) to lymphoid Influence of host age and route of inoculation. J Natl
leukosis. Natl Cancer Inst Monogr. 17:83–98. Cancer Inst. 24:1423–1442.
35 Birmingham, C.L, D. Dupont, P. Riou, C. Armanet, K.N 49 Burmester, B.R, and T.N Fredrickson. 1964.
Edamura, B. Martinho, A. Serres, S. Jacouton, V. Transmission of virus from field cases of avian
Detrez, B. McNeil, M. Schreiber, D. Gaillac, T. lymphomatosis. I. Isolation of virus in line 151
Bonnevay, L. Gisonni‐Lex, and L. Mallet. 2013. chickens. J Natl Cancer Inst. 32:37–63.
Detection of avian retroviruses in vaccines by 50 Burmester, B.R, M.A Gross, W.G Walter, and A.K
amplification on DF‐1 cells with immunostaining and Fontes. 1959. Pathogenicity of a viral strain (RPL 12)
fluorescent product‐enhanced reverse transcriptase causing avian visceral lymphomatosis and related
endpoint methods. J Clin Microbiol. 51:1496–1504. neoplasms. II. Host‐virus interrelations affecting
36 Boni‐Schnetzler, M., J. Boni, F.J Ferdinand, and R.M response. J Natl Cancer Inst. 22:103–127.
Franklin. 1985. Developmental and molecular aspects 51 Burmester, B.R, and N.M Nelson. 1945. The effect of
of nephroblastomas induced by avian myeloblastosis‐ castration and sex hormones upon the incidence of
associated virus 2‐O. J Virol. 55:213–222. lymphomatosis in chickens. Poult Sci. 24.
37 Bova, C.A, J.P Manfredi, and R. 1986. Swanstrom. env 52 Burmester, B.R, and W.G Walter. 1961. Occurrence
genes of avian retroviruses: nucleotide sequence and of visceral lymphomatosis in chickens inoculated
molecular recombinants define host range with Rous sarcoma virus. J Natl Cancer Inst.
determinants. Virology. 152:343–354. 26:511–518.
686 Section II  Viral Diseases

53 Burmester, B.R, W.G Walter, M.A Gross, and A.K major determinant for the induction of lymphoid and
Fontes. 1959. The oncogenic spectrum of two `pure’ myeloid tumours by avian leukosis virus subgroups A
strains of avian leukosis. J Natl Cancer Inst. 23:277–291. and J, respectively. J Gen Virol. 83:2553–2561.
54 Burstein, H., M. Gilead, U. Bendheim, and M. Kotler. 68 Chesters, P.M, L.P Smith, and V. Nair. E 2006. XSR)
1984. Viral aetiology of haemangiosarcoma outbreaks element contributes to the oncogenicity of Avian
among layer hens. Avian Pathol. 13:715–726. leukosis virus (subgroup J). J Gen Virol. 87:2685–2692.
55 Burstein, H., N. Resnick‐Roguel, J. Hamburger, G. Arad, 69 Chi, Y., F. Diaz‐Griffero, C. Wang, J.A Young, and J.
M. Malkinson, and M. Kotler. 1990. Unique sequences Brojatsch. 2002. An NF‐kappa B‐dependent survival
in the env gene of avian hemangioma retrovirus are pathway protects against cell death induced by TVB
responsible for cytotoxicity and endothelial cell receptors for avian leukosis viruses. J Virol. 76:5581–5587.
perturbation. Virology. 179:512–516. 70 Choudat, D., G. Dambrine, B. Delemotte, and F. Coudert.
56 Campbell, J.G 1969. Tumours of the Fowl. William 1996. Occupational exposure to poultry and prevalence
Heinemann Medical Books: London. of antibodies against Marek’s disease virus and avian
57 Campbell, J.G, and E.C Appleby. 1966. Tumours in leukosis retroviruses. Occup Environ Med. 53:403–410.
young chickens bred for rapid body growth (broiler 71 Ciuffi, A. 2016. The benefits of integration. Clinical
chickens). A study of 351 cases. J Pathol Bacteriol. microbiology and infection: the official publication of
92:77–90. the European Society of Clinical Microbiology and
58 Caparini, U. 1896. Fetati leucemici nei polli. Clin Vet Infectious Diseases. 22:324–332.
(Milan). 19:433–435. 72 Clurman, B.E, and W.S Hayward. 1989. Multiple
59 Chai, N., and P. Bates. 2006. Na+/H+ exchanger type 1 proto‐oncogene activations in avian leukosis virus‐
is a receptor for pathogenic subgroup J avian leukosis induced lymphomas: evidence for stage‐specific events.
virus. Proc Natl Acad Sci USA. 103:5531–5536. Mol Cell Biol. 9:2657–2664.
60 Chebloune, Y., J. Rulka, F.L Cosset, S. Valsesia, C. 73 Coffin, J.M, S.H Hughes, and H.E Varmus. (eds.). 1997.
Ronfort, C. Legras, A. Drynda, J. Kuzmak, V.M Nigon, Retroviruses. Cold Spring Harbor Laboratory Press:
and G. Verdier. 1991. Immune response and resistance Cold Spring Harbor, New York.
to Rous sarcoma virus challenge of chickens 74 Collart, K.L, R. Aurigemma, R.E Smith, S. Kawai, and
immunized with cell‐associated glycoproteins provided H.L Robinson. 1990. Infrequent involvement of c‐fos in
with a recombinant avian leukosis virus. J Virol. avian leukosis virus‐induced nephroblastoma. J Virol.
65:5374–5380. 64:3541–3544.
61 Chen, B., W. Pan, L. Zhang, J. Liu, H. Ouyang, Q. Nie, 75 Collins, W.H, W.E Briles, R.M Zsigray, W.R Dunlop, A.C
and X. Zhang. 2014. NHE1 gene associated with avian Corbett, K.K Clark, J.L Marks, and T.P McGrail. 1977.
leukosis virus subgroup J infection in chicken. Mol Biol The B locus (MHC) in the chicken: association with the
Rep 41:6519–6524. fate of RSV‐induced tumors. Immunogenetics. 5:333–343.
62 Chen, M., A.J Granger, M.W Vanbrocklin, W.S Payne, 76 Cooper, M.D, H.G Purchase, D.E Bockman, and W.E
H. Hunt, H. Zhang, J.B Dodgson, and S.L Holmen. Gathings. 1974. Studies on the nature of the
2007. Inhibition of avian leukosis virus replication by abnormality of B cell differentiation in avian lymphoid
vector‐based RNA interference. Virology. 365:464–472. leukosis: production of heterogeneous IgM by tumor
63 Chen, W., Y. Liu, H. Li, S. Chang, D. Shu, H. Zhang, F. cells. J Immunol. 113:1210–1222.
Chen, and Q. Xie. 2015. Intronic deletions of tva receptor 77 Cottral, G.E, B.R Burmester, and N.F Waters. 1954. Egg
gene decrease the susceptibility to infection by avian transmission of avian lymphomatosis. Poult Sci.
sarcoma and leukosis virus subgroup A. Sci Rep. 5:9900. 33:1174–1184.
64 Chen, Y.C, and P.K Vogt. 1977. Endogenous leukosis 78 Crittenden, L.B 1975. Two levels of genetic resistance
viruses in the avian family Phasianidae. Virology. to lymphoid leukosis. Avian Dis. 19:281–292.
76:740–750. 79 Crittenden, L.B 1991. Retroviral elements in the
65 Cheng, J., S. Wen, S. Wang, P. Hao, Z. Cheng, Y. Liu, P. genome of the chickens: Implications for poultry
Zhao, and J. 2017. Liu. gp85 protein vaccine adjuvanted genetics and breeding. Crit Rev Poultry Biol.
with silica nanoparticles against ALV‐J in chickens. 3:73–109.
Vaccine. 35:293–298. 80 Crittenden, L.B, D.A Eagen, and F.A Gulvas. 1979.
66 Chesters, P.M, K. Howes, J.C McKay, L.N Payne, and K. Assays for endogenous and exogenous lymphoid
Venugopal. 2001. Acutely transforming avian leukosis leukosis viruses and chick helper factor with RSV(‐) cell
virus subgroup J strain 966: defective genome encodes lines. Infect Immun. 24:379–386.
a 72‐kilodalton Gag‐Myc fusion protein. J Virol. 81 Crittenden, L.B, and A.M Fadly. 1985. Responses of
75:4219–4225. chickens lacking or expressing endogenous avian
67 Chesters, P.M, K. Howes, L. Petherbridge, S. Evans, L.N leukosis virus genes to infection with exogenous virus.
Payne, and K. Venugopal. 2002. The viral envelope is a Poult Sci. 64:454–463.
Chapter 15  Neoplastic Diseases 687

82 Crittenden, L.B, A.M Fadly, and E.J Smith. 1982. Effect 95 Dai, M., S. Wu, M. Feng, S. Feng, C. Sun, D. Bai, M.
of endogenous leukosis virus genes on response to Gu, M. Liao, and W. Cao. 2016. Recombinant chicken
infection with avian leukosis and reticuloendotheliosis interferon‐alpha inhibits the replication of exogenous
viruses. Avian Dis. 26:279–294. avian leukosis virus (ALV) in DF‐1 cells. Mol
83 Crittenden, L.B, and H.J Kung. 1984. Mechanism of Immunol. 76:62–69.
induction of lymphoid leukosis and related neoplasms 96 de Boer, G.F 1987. Approaches to control of avian
by avian leukosis viruses. In: Mechanisms of Viral lymphoid leukosis. In: Avian Leukosis. G.F de Boer
Leukaemogenesis. J.M Goldman and O. Jarrett (eds.). (ed.). Martinus Nijhoff: Boston, MA. 261–286.
Churchill Livingstone: Edinburgh, Scotland. 64–88. 97 de Boer, G.F, O.J H. Devos, and H.J L. Maas. 1981. The
84 Crittenden, L.B, and J.V Motta. 1969. A survey of incidence of lymphoid leukosis in chickens in the
genetic resistance to leukosis‐sarcoma viruses in Netherlands. Zootechnica Int. 10:32–35.
commercial stocks of chickens. Poult Sci. 98 De The, G., U. Heine, H. Ishiguro, J.R Sommer, D.
48:1751–1757. Beard, and J.W Beard. 1962. Biologic response of
85 Crittenden, L.B, and W. Okazaki. 1966. Genetic nephrogenic cells to avian myeloblastosis virus. Fed
influence of the Rs locus on susceptibility to avian Proc. 21:919–929.
tumor viruses. II. Rous sarcoma virus antibody 99 Di Stefano, H.S, and R.M Dougherty. 1968.
production after strain RPL12 virus inoculation. J Natl Multiplication of avian leukosis virus in the
Cancer Inst. 36:299–303. reproductive system of the rooster. J Natl Cancer Inst.
86 Crittenden, L.B, W. Okazaki, and R. Reamer. Genetic 41:451–464.
1963. Resistance to Rous sarcoma virus in embryo cell 100 Diaz‐Griffero, F., S.A Hoschander, and J. Brojatsch.
cultures and embryos. Virology. 20:541–544. 2003. Bystander killing during avian leukosis virus
87 Crittenden, L.B, W. Okazaki, and E.J Smith. 1983. subgroup B infection requires TVB(S3) signaling.
Incidence of avian leukosis virus infection in broiler J Virol. 77:12552–12561.
stocks and its effect on early growth. Poult Sci. 101 Diaz‐Griffero, F., A.P Jackson, and J. Brojatsch. 2005.
62:2383–2386. Cellular uptake of avian leukosis virus subgroup B is
88 Crittenden, L.B, H.G Purchase, J.J Solomon, W. mediated by clathrin. Virology. 337:45–54.
Okazaki, and B.R Burmester. 1972. Genetic control of 102 DiStefano, H.S, and R.M Dougherty. 1966.
susceptibility to the avian leukosis complex. I. The Mechanisms for congenital transmission of avian
leukosis‐sarcoma virus group. Poult Sci. 51:242–261. leukosis virus. J Natl Cancer Inst. 37:869–883.
89 Crittenden, L.B, and E.J Smith. 1984. A comparison of 103 DiStefano, H.S, and R.M Dougherty. 1968. Multiplication
test materials for differentiating avian leukosis virus of avian leukosis virus in the reproductive system of the
group‐specific antigens of exogenous and endogenous rooster. J Natl Cancer Inst. 41:451–464.
origin. Avian Dis. 28:1057–1070. 104 DiStefano, H.S, and R.M Dougherty. 1969.
90 Crittenden, L.B, E.J Smith, and A.M Fadly. 1984. Multiplication of avian leukosis virus in endocrine
Influence of endogenous viral (ev) gene expression and organs of congenitally infected chickens. J Natl
strain of exogenous avian leukosis virus (ALV) on Cancer Inst. 42:147–154.
mortality and ALV infection and shedding in chickens. 105 Dmochowski, L., C.E Grey, F. Padgett, P.L Langford,
Avian Dis. 28:1037–1056. and B.R Burmester. 1964. Submicroscopic
91 Crittenden, L.B, R.L Witter, and A.M Fadly. 1979. Low morphology of avian neoplasms. VI. Comparative
incidence of lymphoid tumors in chickens studies on Rous sarcoma, visceral lymphomatosis,
continuously producing endogenous virus. Avian Dis. erythroblastosis, myeloblastosis, and nephroblastoma.
23:646–653. Tex Rep Biol Med. 22:20–60.
92 Crittenden, L.B, R.L Witter, W. Okazaki, and P.E 106 Dong, X., F. Meng, T. Hu, S. Ju, Y. Li, P. Sun, Y. Wang,
Neiman. 1979. Lymphoid neoplasms in chicken flocks W. Chen, F. Zhang, H. Su, S. Li, H. Cui, J. Chen, S. Xu,
free of infection with exogenous avian tumor viruses. L. Fang, H. Luan, Z. Zhang, S. Chang, J. Li, L. Wang, P.
J Natl Cancer Inst. 63:191–200. Zhao, W. Shi, and Z. Cui. 2017. Dynamic co‐evolution
93 Cummins, T.J, and R.E Smith. 1988. Analysis of and interaction of avian leukosis virus genetic variants
hematopoietic and lymphopoietic tissue during a and host immune responses. Front Microbiol. 8:1168.
regenerative aplastic crisis induced by avian retrovirus 107 Dong, X., P. Zhao, B. Xu, J. Fan, F. Meng, P. Sun, S. Ju,
MAV‐2(O). Virology. 163:452–461. Y. Li, S. Chang, W. Shi, and Z. Cui. 2015. Avian
94 Dai, M., M. Feng, D. Liu, W. Cao, and M. Liao. 2015. leukosis virus in indigenous chicken breeds, China.
Development and application of SYBR Green I real‐ Emerg Microb Infect. 4:e76.
time PCR assay for the separate detection of subgroup J 108 Dorner, A.J, and J.M Coffin. 1986. Determinants for
Avian leukosis virus and multiplex detection of avian receptor interaction and cell killing on the avian
leukosis virus subgroups A and B. Virol J. 12:52. retrovirus glycoprotein gp85. Cell. 45:365–374.
688 Section II  Viral Diseases

109 Dorner, A.J, J.P Stoye, and J.M Coffin. 1985. Molecular positive and ‐negative chickens exposed to virus at
basis of host range variation in avian retroviruses. J hatching. Avian Dis. 32:89–95.
Virol. 53:32–39. 123 Fadly, A.M 1989. Leukosis and sarcoma. In: A
110 Dou, W., H. Li, Z. Cheng, P. Zhao, J. Liu, Z. Cui, H. Laboratory Manual for the Isolation and Identification
Liu, W. Jing, and H. Guo. 2013. Maternal antibody of Avian Pathogens. H.G Purchase, L.H Arp, C.H
induced by recombinant gp85 protein vaccine Domermuth, J.E Pearson (eds.). American Association
adjuvanted with CpG‐ODN protects against ALV‐J of Avian Pathologists: Kennett Square, PA. 135–142.
early infection in chickens. Vaccine. 31:6144–6149. 124 Fadly, A.M 1992. Some observations on the
111 Dougherty, R.M, and H.S DiStefano. 1967. Sites of enhancement of avian leukosis virus‐induced
avian leukosis virus multiplication in congenitally lymphomas byserotype 2 Marek’s disease virus.
infected chickens. Cancer Res. 27:322–332. Proceedings XIX World’s Poultry Congress. Ponsen &
112 Dougherty, R.M, J.A Stewart, and H.R Morgan. 1960. Looijen: Wageningen. 281–285.
Quantitative studies of the relationships between 125 Fadly, A.M 1993. Induction of antibodies to avian
infecting dose of Rous sarcoma virus, antiviral leukosis and reticuloendotheliosis viruses using
immune response, and tumor growth in chickens. defective retroviral particles. Proceedings, 130th
Virology. 11:349–370. AVMA Convention, Minneapolis, MN.
113 Dren, C.N, and I. Nemeth. 1987. Demonstration of 126 Fadly, A.M 2000. Isolation and identification of avian
immunoglobulin M on avian lymphoid leukosis leukosis viruses: a review. Avian Pathol. 29:529–535.
lymphoma cells by the unlabelled antibody 127 Fadly, A.M, L.B Crittenden, and E.J Smith. 1987.
peroxidase‐antiperoxidase method. Avian Pathol. Variation in tolerance induction and oncogenicity due
16:253–268. to strain of avian leukosis virus. Avian Pathol.
114 Eckert, E.A, D. Beard, and J.W Beard. 1954. Dose‐ 16:665–677.
response relations in experimental transmission of 128 Fadly, A.M, and D.L Ewert. 1994. Enhancement of
avian erythromyeloblastic leukosis III. Titration of the avian retrovirus‐induced B‐cell lymphoma by Marek’s
virus. J Natl Cancer Inst. 14:1055–1066. disease herpesvirus. World Scientific Singapore. 1–9.
115 Elleder, D., V. Stepanets, D.C Melder, F. Senigl, J. 129 Fadly, A.M, L.F Lee, and L.D Bacon. 1982.
Geryk, P. Pajer, J. Plachy, J. Hejnar, J. Svoboda, and M.J Immunocompetence of chickens during early and
Federspiel. 2005. The receptor for the subgroup C tumorigenic stages of Rous‐associated virus‐1
avian sarcoma and leukosis viruses, Tvc, is related to infection. Infect Immun. 37:1156–1161.
mammalian butyrophilins, members of the 130 Fadly, A.M, W. Okazaki, and R.L Witter. 1981.
immunoglobulin superfamily. J Virol. Hatchery‐related contact transmission and short‐
79:10408–10419. term small‐group‐rearing as related to lymphoid‐
116 Ellermann, V., and O. Bang. 1908. Experimentelle leukosis‐virus‐eradication programs. Avian Dis.
leukamie bei huhnern. Zentralbl Bakteriol Parasitenkd 25:667–677.
Infektionskr Hyg Abt I Orig. 46:595–609. 131 Fadly, A.M, R.F Silva, and L.F Lee, eds. 2000. Antigenic
117 Engelman, A.N, and P. Cherepanov. 2017. Retroviral characterisation of selected field isolates of subgroup J
intasomes arising. Curr Op Struct Biol. 47:23–29. avian leukosis virus. Rauischholzhausen, Germany.
118 Enrietto, P., and M. Hayman. 1987. Structure and 132 Fadly, A.M, and E.J Smith. 1999. Isolation and some
virus‐associated oncogenes of avian sarcoma and characteristics of a subgroup J‐like avian leukosis
leukaemia viruses. In: Avian Leukosis. G.F De Boer, virus associated with myeloid leukosis in meat‐type
ed. Martinus Nijhoff, Boston. 29–46. chickens in the United States. Avian Dis. 43:391–400.
119 Enrietto, P.J, M.J Hayman, G.M Ramsay, J.A Wyke, 133 Fadly, A.M, and R.L Witter. 1993. Effects of age at
and L.N Payne. 1983. Altered pathogenicity of avian infection with serotype 2 Marek’s disease virus on
myelocytomatosis (MC29) viruses with mutations in enhancement of avian leukosis virus‐induced
the v‐myc gene. Virology. 124:164–172. lymphomas. Avian Pathol. 22:565–576.
120 Ewert, D.L, I. Steiner, and J. Duttadaway. 1990. In ovo 134 Fadly, A.M, and R.L Witter. 1998. Oncornaviruses:
infection with the avian retrovirus RAV‐1 leads to Leukosis/Sarcoma and reticuloendotheliosis. In: A
persistent infection of the central nervous system. Lab Laboratory Manual for the isolation and identification
Invest. 62:156–116. of Avian Pathogens, 4th ed. J.R Glisson, D.J Jackwood,
121 Fadly, A., R. Silva, H. Hunt, A. Pandiri, and C. Davis. J.E Pearson, W.M Reed, and D.E Swayne (eds.). Am.
2006. Isolation and characterization of an adventitious Assoc. Avian Pathologists: Kennet Square, PA 185–196.
avian leukosis virus isolated from commercial Marek’s 135 Fadly, A.M, R.L Witter, and L.F Lee. 1985. Effects of
disease vaccines. Avian Dis. 50:380–385. chemically or virus‐induced immunodepression on
122 Fadly, A.M 1988. Avian leukosis virus (ALV) infection, response of chickens to avian leukosis virus. Avian
shedding, and tumors in maternal ALV antibody‐ Dis. 29:12–25.
Chapter 15  Neoplastic Diseases 689

136 Feng, M., M. Dai, T. Xie, Z. Li, M. Shi, and X. Zhang. PCR method for rapid differential detection of
2016. Innate immune responses in ALV‐J infected subgroup A, B, and J avian leukosis viruses. J Clin
chicks and chickens with hemangioma in vivo. Front Microbiol. 52:37–44.
Microbiol. 7:786. 150 Garcia‐Fernandez, R.A, C. Perez‐Martinez, J.
137 Feng, M., and X. Zhang. 2016. Immunity to avian Espinosa‐Alvarez, A. Escudero‐Diez, J.F Garcia‐
leukosis virus: where are we now and what should we Marin, A. Nunez, and M.J Garcia‐Iglesias. 2000.
do? Front Immunol. 7:624. Lymphoid leukosis in an ostrich (Struthio camelus).
138 Feng, W., W. Meng, L. Cai, X. Cui, Z. Pan, G. Wang, Vet Rec. 146:676–677.
and Z. Cheng. 2016. Avian leukosis virus subgroup J 151 Garcia, M., J. El‐Attrache, S.M Riblet, V.R Lunge, A.S
induces its receptor–chNHE1 up‐regulation. Virol J. Fonseca, P. Villegas, and N. Ikuta. 2003. Development
13:58. and application of reverse transcriptase nested
139 Feng, W., D. Zhou, W. Meng, G. Li, P. Zhuang, Z. Pan, polymerase chain reaction test for the detection of
G. Wang, and Z. Cheng. 2017. Growth retardation exogenous avian leukosis virus. Avian Dis. 47:41–53.
induced by avian leukosis virus subgroup J associated 152 Gavora, J.S 1987. Influences of avian leukosis virus
with down‐regulated Wnt/beta‐catenin pathway. infection on production and mortality and the role of
Microbial Pathog. 104:48–55. genetic selection in the control of lymphoid leukosis.
140 Foster, R.G, J.B Lian, G. Stein, and H.L Robinson. In: Avian Leukosis. G.F de Boer (ed.). Martinus
1994. Replication of an osteopetrosis‐inducing avian Nijhoff: Boston, MA. 241–260.
leukosis virus in fibroblasts, osteoblasts, and 153 Gavora, J.S, J.L Spencer, and J.A Chambers. 1982.
osteopetrotic bone. Virology. 205:179–187. Performance of meat‐type chickens test‐positive and
141 Frank, R.M, and R.M Franklin. 1982. Electron ‐negative for lymphoid leukosis virus infection. Avian
microscopy of avian osteopetrosis induced by Pathol. 11:29–38.
retrovirus MAV2‐0. Calcif Tissue Int. 34:382–390. 154 Gavora, J.S, J.L Spencer, R.S Gowe, and D.L Harris.
142 Franklin, R.M, and M.T Martin. 1980. In ovo 1980. Lymphoid leukosis virus infection: effects on
tumorigenesis induced by avian osteopetrosis virus. production and mortality and consequences in
Virology. 105:245–249. selection for high egg production. Poult Sci. 59:2165–2178.
143 Fredrickson, T.N, B.R Burmester, and W. Okazaki. 155 Gebriel, G.M, and A.W Nordskog. 1983. Genetic
1965. Transmission of virus from field cases of avian linkage of subgroup C Rous sarcoma virus‐induced
lymphomatosis. II. Development of strains by serial tumour expression in chickens to the IR‐GAT locus of
passage in line 15I chickens. Avian Dis. 9:82–103. the B complex. J Immunogenet. 10:231–235.
144 Fredrickson, T.N, H.G Purchase, and B.R Burmester. 156 Gilka, F., and J.L Spencer. 1985. Viral matrix inclusion
1964. Transmission of virus from field cases of avian bodies in myocardium of lymphoid leukosis virus‐
lymphomatosis. III. Variation in the oncogenic spectra infected chickens. Am J Vet Res. 46:1953–1960.
of passaged virus isolates. Natl Cancer Inst Monogr. 157 Gilka, F., and J.L Spencer. 1987. Importance of the
17:1–29. medullary macrophage in the replication of lymphoid
145 Frisby, D., R. MacCormick, and R. Weiss. 1980. Origin leukosis virus in the bursa of Fabricius of chickens.
of RAV‐0. The endogenous retrovirus of chickens. Am J Vet Res. 48:613–620.
Cold Spring Harb Conf Cell Prolifer. 7:509–517. 158 Gilka, F., and J.L Spencer. 1990. Chronic myocarditis
146 Fung, Y.K, L.B Crittenden, A.M Fadly, and H.J Kung. and circulatory syndrome in a White Leghorn strain
1983. Tumor induction by direct injection of cloned induced by an avian leukosis virus: light and electron
v‐src DNA into chickens. Proc Natl Acad Sci USA. microscopic study. Avian Dis. 34:174–184.
80:353–357. 159 Gingerich, E., R.E Porter, B. Lupiani, and A.M Fadly.
147 Fung, Y.K, W.G Lewis, L.B Crittenden, and H.J Kung. 2002. Diagnosis of myeloid leukosis induced by a
1983. Activation of the cellular oncogene c‐erbB by recombinant avian leukosis virus in commercial white
LTR insertion: molecular basis for induction of leghorn egg laying flocks. Avian Dis. 46:745–748.
erythroblastosis by avian leukosis virus. Cell. 160 Gong, M., H.L Semus, K.J Bird, B.J Stramer, and A.
33:357–368. Ruddell. 1998. Differential selection of cells with
148 Fynan, E., T.M Block, J. DuHadaway, W. Olson, and proviral c‐myc and c‐erbB integrations after avian
D.L Ewert. 1992. Persistence of Marek’s disease virus leukosis virus infection. J Virol. 72:5517–5525.
in a subpopulation of B cells that is transformed by 161 Goodwin, M.A, S. Hafner, D.I Bounous, and J. Brown.
avian leukosis virus, but not in normal bursal B cells. 1998. Myeloid leukosis is a misnomer: multiple and
J Virol. 66:5860–5866. diverse tumor morphotypes are found in broiler
149 Gao, Q., B. Yun, Q. Wang, L. Jiang, H. Zhu, Y. Gao, L. breeder chickens that are infected with subgroup J
Qin, Y. Wang, X. Qi, H. Gao, X. Wang, and Y. Gao. avian leukosis/sarcoma viruses (J‐ALSV). In:
2014. Development and application of a multiplex Proceedings, 135th AVMA Convention. Baltimore. 189.
690 Section II  Viral Diseases

162 Goubin, G., D.S Goldman, J. Luce, P.E Neiman, and 177 Hayward, W.S, and B.G Neel. 1981. Retroviral gene
G.M Cooper. 1983. Molecular cloning and nucleotide expression. Curr Top Microbiol Immunol. 19:217–276.
sequence of a transforming gene detected by 178 Heine, U., G. De The, H. Ishiguro, J.R Sommer, D.
transfection of chicken B‐cell lymphoma DNA. Beard, and J.W Beard. 1962. Multiplicity of cell
Nature. 302:114–119. response to the BAI strain A (myeloblastosis) avian
163 Graf, T. 1972. A plaque assay for avian RNA tumor tumor virus. II. Nephroblastoma (Wilms’ tumor):
viruses. Virology. 50:567–578. ultrastructure. J Natl Cancer Inst. 29:41–105.
164 Graf, T. 1975. In vitro transformation of chicken 179 Hirota, Y., M.T Martin, M. Viljanen, P. Toivanen, and
bone marrow cells with avian erythroblastosis virus. R.M Franklin. 1980. Immunopathology of chickens
Z Naturforsch [C]. 30:847–849. infected in ovo and at hatching with the avian
165 Graf, T., and H. Beug. 1978. Avian leukemia viruses: osteopetrosis virus MAV.2–0. Eur J Immunol.
interaction with their target cells in vivo and in vitro. 10:929–936.
Biochim Biophys Acta. 516:269–299. 180 Hofmann, A., J. Plachy, L. Hunt, J. Kaufman, and K.
166 Grawenhoff, J., and A.N Engelman. 2017. Retroviral 2003. Hala. v‐src oncogene‐specific carboxy‐terminal
integrase protein and intasome nucleoprotein peptide is immunoprotective against Rous sarcoma
complex structures. World J Biol Chem. 8:32–44. growth in chickens with MHC class I allele B‐F12.
167 Gray, E.R, C.J Illingworth, J.M Coffin, and J.P Stoye. Vaccine. 21:4694–4699.
2011. Binding of more than one Tva800 molecule is 181 Holmes, J.R 1964. Avian osteopetrosis. Natl Cancer
required for ASLV‐A entry. Retrovirol. 8:96. Inst Monogr. 17:63–79.
168 Gross, M.A, B.R Burmester, and W.G Walter. 1959. 182 Humphries, E.H, and T.W Baba. 1984. Follicular
Pathogenicity of a viral strain (RPL12) causing avian hyperplasia in the prelymphomatous avian bursa:
visceral lymphomatosis and related neoplasms. I. relationship to the incidence of B‐cell lymphomas.
Nature of the lesions. J Natl Cancer Inst. 22:83–101. Curr Top Microbiol Immunol. 113:47–55.
169 Hafner, S., M.A Goodwin, E.J Smith, D.I Bounous, M. 183 Humphries, E.H, and T.W Baba. 1986. Restrictions
Puette, L.C Kelley, K.A Langheinrich, and A.M Fadly. that influence avian leukosis virus‐induced lymphoid
1996. Multicentric histiocytosis in young chickens. leukosis. Curr Top Microbiol Immunol. 132:215–220.
Gross and light microscopic pathology. Avian Dis. 184 Hunt, H., A. Fadly, R. Silva, and H. Zhang. 2008.
40:202–209. Survey of endogenous virus and TVB* receptor status
170 Hafner, S., M.A Goodwin, E.J Smith, A. Fadly, and L.C of commercial chicken stocks supplying specific‐
Kelley. 1998. Pulmonary sarcomas in a young chicken. pathogen‐free eggs. Avian Dis. 52:433–440.
Avian Dis. 42:824–828. 185 Hunt, H., B. Lupiani, and A.M Fadly. 2000.
171 Hafner, S., S.M Williams, and M.T Sutton. 2007. Recombination between ALV‐J and endogenous
Retroviral inclusions in the enteric smooth muscle of subgroup E viruses. In: Proceedings, International
a tumor‐bearing young chicken. Avian Dis. 51:133–136. Symposium on ALV‐J and Other Avian Retroviruses.
172 Haguenau, F., and J.W Beard. 1962. The avian E.F Kaleta, L.N Payne, and U. Heffels‐Redmann (eds).
sarcoma‐leukosis complex: its biology and Rauischholzhausen, Germany. 50–60.
ultrastructure. In: Tumors Induced by Viruses. A.J 186 Hunt, H.D, L.F Lee, D. Foster, R.F Silva, and A.M
Dalton and F. Haguenau, eds. Academic Press, Fadly. 1999. A genetically engineered cell line resistant
New York. 1–59. to subgroup J avian leukosis virus infection (C/J).
173 Haguenau, F., H. Febvre, and J. Arnoult. 1960. Virology. 264:205–210.
Ultrastructure of Rous sarcoma virus cultivated in 187 Hussain, A.I, J.A Johnson, M. Da Silva Freire, and W.
vitro. C R Hebd Seances Acad Sci. 250:1747–1749. Heneine. 2003. Identification and characterization of
174 Han, J.Y, and B.R Lee. 2017. Isolation and avian retroviruses in chicken embryo‐derived yellow
characterization of chicken primordial germ cells and fever vaccines: investigation of transmission to
their application in transgenesis. Methods Mol Biol. vaccine recipients. J Virol. 77:1105–1111.
1650:229–242. 188 Hussain, A.I, V. Shanmugam, W.M Switzer, S.X Tsang,
175 Harris, D.L, V.A Garwood, P.C Lowe, P.Y Hester, L.B A. Fadly, D. Thea, R. Helfand, W. Bellini, T.M Folks,
Crittenden, and A.M Fadly. 1984. Influence of sex‐ and W. Heneine. 2001. Lack of evidence of
linked feathering phenotypes of parents and progeny endogenous avian leukosis virus and endogenous
upon lymphoid leukosis virus infection status and egg avian retrovirus transmission to measles, mumps and
production. Poult Sci. 63:401–413. rubella vaccine recipients. Emerg Infect Dis. 7:66–72.
176 Hauptli, D., L. Bruckner, and H.P Ottiger. 1997. Use of 189 Ignjatovic, J., R.A Fraser, and T.J Bagust. 1986. Effect
reverse transcriptase polymerase chain reaction for of lymphoid leukosis virus on performance of layer
detection of vaccine contamination by avian leukosis hens and the identification of infected chickens by
virus. J Virol. Methods 66:71–81. tests on meconia. Avian Pathol. 15:63—74.
Chapter 15  Neoplastic Diseases 691

190 Ishiguro, H., D. Beard, J.R Sommer, U. Heine, d. Thé., 203 Kirev, T.T, I.A Toshkov, and Z.M Mladenov. 1986.
and J.W Beard. 1962. Multiplicity of cell response to Virus‐induced pancreatic cancer in guinea fowl: a
the BAI strain A (myeloblastosis) avian tumor virus. I. morphologic study. J Natl Cancer Inst. 77:713–720.
Nephroblastoma (Wilms’ tumor): Gross and 204 Kirev, T.T, T.A Toshkov, and Z.M Mladenov. 1987.
microscopic pathology. J Natl Cancer Inst. 29:1–39. Virus‐induced duodenal adenomas in guinea fowl.
191 Ishizaki, R., A.J Langlois, and D.P Bolognesi. 1975. J Natl Cancer Inst. 79:1117–1121.
Isolation of two subgroup‐specific leukemogenic 205 Klucking, S., and J.A Young. 2004. Amino acid
viruses from standard avian myeloblastosis virus. residues Tyr‐67, Asn‐72, and Asp‐73 of the TVB
J Virol. 15:906–912. receptor are important for subgroup E avian sarcoma
192 Johnson, E.S, Y. Zhou, C.L Yau, V. Sarda, N. Preacely, and leukosis virus interaction. Virology. 318:371–380.
S. Bankuru, S. Bangara, M. Felini, and H. Ndetan. 206 Kucerova, D., J. Plachy, M. Reinisova, F. Senigl, K.
2011. Update of cancer and non‐cancer mortality in Trejbalova, J. Geryk, and J. Hejnar. 2013.
the Missouri poultry cohort. Am J Ind Med. 54:49–54. Nonconserved tryptophan 38 of the cell surface
193 Joliot, V., K. Boroughs, F. Lasserre, J. Crochet, G. receptor for subgroup J avian leukosis virus
Dambrine, R.E Smith, and B. Perbal. 1993. Pathogenic discriminates sensitive from resistant avian species.
potential of myeloblastosis‐associated virus: J Virol. 87:8399–8407.
implication of env proteins for osteopetrosis 207 Kumanishi, T. 1967. Brain tumors induced with Rous
induction. Virology. 195:812–819. sarcoma virus, Schmidt‐Ruppin strain. I. Induction of
194 Joliot, V., C. Martinerie, G. Dambrine, G. Plassiart, M. brain tumors in adult mice with Rous chicken sarcoma
Brisac, J. Crochet, and B. Perbal. 1992. Proviral cells. Jpn J Exp Med. 37:461–474.
rearrangements and overexpression of a new cellular 208 Kumanishi, T., F. Ikuta, K. Nishida, K. Ueki, and T.
gene (nov) in myeloblastosis‐associated virus type 1‐ Yamamoto. 1973. Brain tumors induced in adult
induced nephroblastomas. Mol Cell Biol. 12:10–21. monkeys by Schmidt‐Ruppin strain of Rous sarcoma
195 Justice, J. F. t., R.W Morgan, and K.L Beemon. 2015. virus. Gann. 64:641–643.
Common Viral Integration Sites Identified in Avian 209 Kumanishi, T., F. Ikuta, and T. Yamamoto. 1973. Brain
Leukosis Virus‐Induced B‐Cell Lymphomas. mBio. tumors induced by Rous sarcoma virus, Schmidt‐
6:e01863–01815. Ruppin strain. 3. Morphology of brain tumors induced
196 Justice, J. t., S. Malhotra, M. Ruano, Y. Li, G. Zavala, in adult mice. J Natl Cancer Inst. 50:95–109.
N. Lee, R. Morgan, and K. Beemon. 2015. The MET 210 Kumanishi, T., and T. Yamamoto. 1970. Brain tumors
gene is a common integration target in avian leukosis induced with Rous sarcoma virus, Schmidt‐Ruppin
virus subgroup J‐induced chicken hemangiomas. strain. 2. Rous tumor specific transplantation antigen
J Virol. 89:4712–4719. in subcutaneously passaged mouse brain tumors. Jpn J
197 Ka, S., S. Kerje, L. Bornold, U. Liljegren, P.B Siegel, L. Exp Med. 40:79–86.
Andersson, and F. Hallbook. 2009. Proviral 211 Kung, H.J, and J.L Liu. 1997. Retroviral oncogenesis.
integrations and expression of endogenous avian In: Viral Pathogenesis. N. Nathanson, ed. Lippincott‐
leucosis virus during long term selection for high and Raven Publishers, Philadelphia. 235–266.
low body weight in two chicken lines. Retrovirol. 6:68. 212 Kurth, R., and H. Bauer. 1972. Cell‐surface antigens
198 Kanter, M.R, R.E Smith, and W.S Hayward. 1988. induced by avian RNA tumor viruses: Detection by a
Rapid induction of B‐cell lymphomas: insertional cytotoxic microassay. Virology. 47:426–433.
activation of c‐myb by avian leukosis virus. J Virol. 213 Kurth, R., E.M Fenyo, E. Klein, and M. Essex. 1979.
62:1423–1432. Cell‐surface antigens induced by RNA tumour
199 Kelloff, G., M. Hatanaka, and R.V Gilden. 1972. Assay viruses. Nature. 279:197–201.
of C‐type virus infectivity by measurement of RNA‐ 214 Labat, M.L 1986. Retroviruses, Immunosuppression
dependent DNA polymerase activity. Virology. and Osteopetrosis. Biomed Pharmacother 40:85–90.
48:266–269. 215 Lai, H., H. Zhang, Z. Ning, R. Chen, W. Zhang, A.
200 Kelloff, G., and P.K Vogt. 1966. Localization of avian Qing, C. Xin, K. Yu, W. Cao, and M. Liao. 2011.
tumor virus group‐specific antigen in cell and virus. Isolation and characterization of emerging subgroup J
Virology. 29:377–384. avian leukosis virus associated with hemangioma in
201 King, A.M Q., M.J Adams, E.B Carstens, and L.E J. egg‐type chickens. Vet Microbiol. 151:275–283.
Virus Taxonomy: IXth 2012. Report of the 216 Lapis, K. 1979. Histology and ultrastructural aspects
International Committee on Taxonomy of Viruses. of virus‐induced primary liver cancer and
Elsevier‐Academic Press, London‐San Diego. transplantable hepatomas of viral origin in chickens.
202 Kirev, T.T 1988. Neoplastic response of guinea fowl to J Toxicol Environ Health. 5:469–501.
osteopetrosis virus strain MAV‐2(0). Avian Pathol. 217 Lee, H.J, K.Y Lee, K.M Jung, K.J Park, K.O Lee, J.Y
17:101–112. Suh, Y. Yao, V. Nair, and J.Y Han. 2017. Precise gene
692 Section II  Viral Diseases

editing of chicken Na+/H+ exchange type 1 (chNHE1) epidemiology of J‐subgroup avian leukosis virus
confers resistance to avian leukosis virus subgroup J isolated from meat‐type chickens in southern
(ALV‐J). Dev Comp Immunol. 77:340–349. China between 2013 and 2014. Arch Virol.
218 Lee, H.J, K.Y Lee, Y.H Park, H.J Choi, Y. Yao, V. Nair, 161:3039–3046.
and J.Y Han. 2017. Acquisition of resistance to avian 230 Liu, D., M. Dai, X. Zhang, W. Cao, and M. Liao.
leukosis virus subgroup B through mutations on tvb Subgroup 2016. J avian leukosis virus infection of
cysteine‐rich domains in DF‐1 chicken fibroblasts. Vet chicken dendritic cells induces apoptosis via the
Res. 48:48. aberrant expression of microRNAs. Sci Rep. 6:20188.
219 Lee, J., S. Mun, D.H Kim, C.S Cho, D.Y Oh, and K. 231 Liu, D., Q. Qiu, X. Zhang, M. Dai, J. Qin, J. Hao, M.
Han. 2017. Chicken (Gallus gallus) endogenous Liao, and W. Cao. 2016. Infection of chicken bone
retrovirus generates genomic variations in the chicken marrow mononuclear cells with subgroup J avian
genome. MobileDNA. 8:2. leukosis virus inhibits dendritic cell differentiation
220 Lee, L.F, A.M Fadly, and H.D Hunt. 2000. Avian and alters cytokine expression. Infect Genet Evol.
leukosis virus subgroup J envelope gene product for 44:130–136.
diagnosis and immunogenic composition. United 232 Lounkova, A., J. Kosla, D. Prikryl, K. Stafl, D.
States Patent #6:146,641. Kucerova, and J. Svoboda. 2017. Retroviral host range
221 Lee, R.M, G. Gillet, and P. Neiman. 1998. Molecular extension is coupled with Env‐activating mutations
events in avian neoplasia: regulation of cell death in resulting in receptor‐independent entry. Proc Natl
development of B‐cell lymphomas in the chicken Acad Sci USA. 114:E5148–e5157.
bursa of Fabricius. Avian Pathol. 27:S16–S20. 233 Lupiani, B., Arun Pandiri, Jody Mays, Henry Hunt,
222 Lesbats, P., A.N Engelman, and P. Cherepanov. 2016. and Aly Fadly 2006. Molecular and biological
Retroviral DNA integration. Chem Rev. characterization of a naturally occurring recombinant
116:12730–12757. subgroup B avian leukosis virus (ALV) with a
223 Li, N., B. Xu, W. Dong, S. Qiao, L.F Lee, H.M Zhang, subgroup J like long terminal repeat (LTR). Avian Dis.
M. Li, and N. Du. 2007. Detection and localization of 50: In Press.
naturally transmitted avian leukosis subgroup J virus 234 Lupiani, B., H. Hunt, R. Silva, and A. Fadly. 2000.
in egg‐type chickens by in situ PCR hybridization. Identification and characterization of recombinant
J Vet Med A Physiol Pathol Clin Med. 54:553–558. subgroup J avian leukosis viruses (ALV) expressing
224 Li, X., W. Lin, S. Chang, P. Zhao, X. Zhang, Y. Liu, W. subgroup A ALV envelope. Virology. 276:37–43.
Chen, B. Li, D. Shu, H. Zhang, F. Chen, and Q. Xie. 235 Lupiani, B., S.M Williams, R.F Silva, H.D Hunt, and
2016. Isolation, identification and evolution analysis A.M Fadly. 2003. Pathogenicity of two recombinant
of a novel subgroup of avian leukosis virus isolated avian leukosis viruses. Avian Dis. 47:425–432.
from a local Chinese yellow broiler in South China. 236 Ma, Y.K, and A.S Khan. 2009. Evaluation of different
Arch Virol. 161:2717–2725. RT enzyme standards for quantitation of retroviruses
225 Li, X., Q. Wang, Y. Gao, X. Qi, Y. Wang, H. Gao, Y. using the single‐tube fluorescent product‐enhanced
Gao, and X. Wang. 2015. Quantitative iTRAQ reverse transcriptase assay. J Virol Methods.
LC‐MS/MS proteomics reveals the proteome profiles 157:133–140.
of DF‐1 cells after infection with subgroup J avian 237 Maas, H.J L., G. F. d. Boer, and J.E Groenendal. 1982.
leukosis virus. BioMed Res Int. 2015:395307. Age related resistance to avian leukosis virus. III.
226 Li, Y., S. Cui, W. Li, Y. Wang, Z. Cui, P. Zhao, and S. Infectious virus, neutralising antibody, and tumours in
Chang. 2017. Vertical transmission of avian leukosis chickens inoculated at various ages. Avian Pathol.
virus subgroup J (ALV‐J) from hens infected through 11:309–327.
artificial insemination with ALV‐J infected semen. 238 Maeda, N., H. Fan, and Y. Yoshikai. 2008. Oncogenesis
BMC Vet Res. 13:204. by retroviruses: old and new paradigms. Rev Med
227 Li, Y., J. Fu, S. Cui, F. Meng, Z. Cui, J. Fan, S. Chang, Virol. 18:387–405.
and P. Zhao. 2017. Gp85 genetic diversity of avian 239 Malfavon‐Borja, R., and C. Feschotte. 2015. Fighting
leukosis virus subgroup J among different individual fire with fire: endogenous retrovirus envelopes as
chickens from a native flock. Poult Sci. 96:1100–1107. restriction factors. J Virol. 89:4047–4050.
228 Liao, C.T, S.Y Chen, W.G Chen, Y. Liu, B.L Sun, H.X 240 Marsh, J.D, L.D Bacon, and A.M Fadly. 1995. Effect of
Li, H.M Zhang, H. Qu, J. Wang, D.M Shu, and Q.M serotype 2 and 3 Marek’s disease vaccines on the
Xie. 2014. Single nucleotide polymorphism variants development of avian leukosis virus‐induced pre‐
within tva and tvb receptor genes in Chinese chickens. neoplastic bursal follicles. Avian Dis. 39:743–751.
Poult Sci. 93:2482–2489. 241 Masegi, T., Y. Inoue, T. Yanai, and K. Ueda. 1993. An
229 Lin, W., X. Li, Z. Dai, X. Zhang, S. Chang, P. Zhao, ultrastructural study of cutaneous hemangioma in two
H. Zhang, F. Chen, and Q. Xie. 2016. Molecular chickens. J Vet Med Sci. 55:185–188.
Chapter 15  Neoplastic Diseases 693

242 McBride, A.A The 2017. Promise of proteomics in the 256 Moscovici, C. 1975. Leukemic transformation with
study of oncogenic viruses. Mol Cell Proteomics. avian myeloblastosis virus: present status. Curr Top
16:S65–s74. Microbiol Immunol. 71:79–101.
243 McBride, M.A T., and R.M Shuman. 1988. Immune 257 Moscovici, C., and L. Gazzolo. 1987. Virus‐cell
response of chickens inoculated with a recombinant interactions of avian sarcoma and defective leukemia
avian leukosis virus. Avian Dis. 32:96–102. viruses. In: Avian Leukosis. G.F De Boer, ed. Martinus
244 Melder, D.C, G.M Pike, M.W VanBrocklin, and M.J Nijhoff, Boston. 153–170.
Federspiel. 2015. Model of the TVA receptor 258 Moscovici, C., L. Gazzolo, and M.G Moscovici. 1975.
determinants required for efficient infection by Focus assay and defectiveness of avian myeloblastosis
subgroup A avian sarcoma and leukosis viruses. virus. Virology. 68:173–181.
J Virol. 89:2136–2148. 259 Moscovici, M.G, and C. Moscovici. 1980. AMV‐
245 Meng, F., X. Dong, T. Hu, S. Chang, J. Fan, P. Zhao, induced transformation of hemopoietic cells: Growth
and Z. Cui. 2016. A deep sequencing reveals patterns of producers and nonproducers. In: In Vivo
significant diversity among dominant variants and and In Vitro Erythropoiesis: The Friend System. G.B
evolutionary dynamics of avian leukosis viruses in two Rossi (ed.). Elsevier/North Holland Biomedical Press:
infectious ecosystems. BMC Vet Res. 12:287. Amsterdam, The Netherlands. 503–514.
246 Meng, F., X. Li, J. Fang, Y. Gao, L. Zhu, G. Xing, F. 260 Motta, J.V, L.B Crittenden, and W.O Pollard. 1973.
Tian, Y. Gao, X. Dong, S. Chang, P. Zhao, Z. Cui, and The inheritance of resistance to subgroup C leukosis‐
Z. Liu. 2016. Genomic diversity of the avian leukosis sarcoma viruses in New Hampshire chickens. Poult
virus subgroup J gp85 gene in different organs of an Sci. 52:578–586.
infected chicken. J Vet Sci. 17:497–503. 261 Motta, J.V, L.B Crittenden, H.G Purchase, H.A Stone,
247 Meng, Q.W, Z.P Zhang, W. Wang, J. Tian, and Z.G and R.L Witter. 1975. Low oncogenic potential of
Xiao. 2011. Enhanced inhibition of Avian leukosis avian endogenous RNA tumor virus infection or
virus subgroup J replication by multi‐target miRNAs. expression. J Natl Cancer Inst. 55:685–689.
Virol J. 8:556. 262 Mucksova, J., J. Plachy, O. Stanek, J. Hejnar, J. Kalina,
248 Meyers, P. 1976. Antibody response to related leukosis B. Benesova, and P. Trefil. 2017. Cytokine response to
viruses induced in chickens tolerant to an avian the RSV antigen delivered by dendritic cell‐directed
leukosis virus. J Natl Cancer Inst. 56:381–386. vaccination in congenic chicken lines. Vet Res. 48:18.
249 Meyers, P., and R.M Dougherty. 1972. Analysis of 263 Munguia, A., and M.J Federspiel. 2008. Efficient
immunoglobulins in chicken antibody to avian subgroup C avian sarcoma and leukosis virus receptor
leucosis viruses. Immunol. 23:1–6. activity requires the IgV domain of the Tvc receptor
250 Mizuno, Y., and K. Arai. 1981. Assay of avian leukosis and proper display on the cell membrane. J Virol.
viruses by indirect immunoperoxidase method. Natl 82:11419–11428.
Inst Anim Health Q (Tokyo). 21:63–67. 264 Murase, A., N. Tamura, N. Matsui, and M. Nakamura.
251 Mizuno, Y., and H. Hatakeyama. 1983. Detection of 1997. Histopathological studies on hemangioma in
antibodies against avian leukosis viruses with indirect broilers at meat inspection. J Jpn Soc Poult Dis.
immunoperoxidase absorbance test. Nippon Juigaku 33:228–232.
Zasshi. 45:31–37. 265 Nair, V. 2013. Tunors of the avian immune system. In:
252 Mizuno, Y., and S. Itohara. 1986. Enzyme‐linked Avian Immunology, 2nd ed. K.A Schat, B. Kaspers and
immunosorbent assay to detect subgroup‐specific P. Kaiser, eds. 333–344.
antibodies to avian leukosis viruses. Am J Vet Res. 266 Nair, V., and A. Fadly. 2013. Leukosis/Sarcoma group.
47:551–556. In: Diseases of Poultry, 13th ed. D.E Swayne, J.R
253 Mladenov, Z., U. Heine, D. Beard, and J.W Beard. Glisson, L.R McDougald, L.K Nolan, D.L Suarez and
Strain 1967. MC29 avian leukosis virus. Myelocytoma, V. Nair, eds. John Wiley & Sons, Inc., Ames, Iowa.
endothelioma, and renal growths: pathomorphological 553–592.
and ultrastructural aspects. J Natl Cancer Inst. 267 Nakamura, K., F. Abe, H. Hihara, and T. Taniguchi.
38:251–285. 1988. Myocardial cytoplasmic inclusions in chickens
254 Mladenov, Z., S. Nedyalkov, I. Ivanov, and I. Toshkov. with hemangioma and lymphoid leukosis. Avian
1980. Neoplastic growths in chickens treated with cell Pathol. 17:3–10.
and cell‐free material from transplantable hepatoma 268 Nakamura, K., M. Ogiso, K. Tsukamoto, N. Hamazaki,
induced by virus strain MC‐29. Neoplasma. 27:175–182. H. Hihara, and N. Yuasa. 2000. Lesions of bone and
255 Moloney, J.B 1956. Biological studies on the Rous bone marrow in myeloid leukosis occurring naturally
sarcoma virus. V. Preparation of improved standard in adult broiler breeders. Avian Dis. 44:215–221.
lots of the virus for use in quantitative investigations. 269 Nakamura, S., K. Ochiai, A. Abe, S. Kishi, K.
J Natl Cancer Inst. 16:877–888. Takayama, and Y. Sunden. 2014. Astrocytic growth
694 Section II  Viral Diseases

through the autocrine/paracrine production of 284 Okazaki, W., R.L Witter, C. Romero, K. Nazerian, J.M
IL‐1beta in the early infectious phase of fowl glioma‐ Sharma, A.M Fadly, and D. Ewert. 1980. Induction of
inducing virus. Avian Pathol. 43:437–442. lymphoid leukosis transplantable tumours and the
270 Nakamura, S., K. Ochiai, H. Hatai, A. Ochi, Y. Sunden, establishment of lymphoblastoid cell lines. Avian
and T. Umemura. 2011. Pathogenicity of avian Pathol. 9:311–329.
leukosis viruses related to fowl glioma‐inducing virus. 285 Pajer, P., V. Pecenka, V. Karafiat, J. Kralova, Z. Horejsi,
Avian Pathol. 40:499–505. and M. Dvorak. 2003. The twist gene is a common
271 Nakamura, Y. 2017. Avian biotechnology. Adv Exp target of retroviral integration and transcriptional
Med Biol. 1001:187–214. deregulation in experimental nephroblastoma.
272 Nehyba, J., J. Svoboda, I. Karakoz, J. Geryk, and J. Oncogene. 22:665–673.
Hejnar. 1990. Ducks: a new experimental host system 286 Pajer, P., V. Pecenka, J. Kralova, V. Karafiat, D.
for studying persistent infection with avian leukaemia Prukova, Z. Zemanova, R. Kodet, and M. Dvorak.
retroviruses. J Gen Virol. 71(9):1937–1945. 2006. Identification of potential human oncogenes by
273 Neiman, P. The 1985. Blym oncogenes. Adv Cancer mapping the common viral integration sites in avian
Res. 45:107–123. nephroblastoma. Cancer Res. 66:78–86.
274 Neiman, P.E, K. Elsaesser, G. Loring, and R. Kimmel. 287 Pan, W., Y. Gao, F. Sun, L. Qin, Z. Liu, B. Yun, Y.
2008. Myc oncogene‐induced genomic instability: Wang, X. Qi, H. Gao, and X. Wang. 2011. Novel
DNA palindromes in bursal lymphomagenesis. PLoS sequences of subgroup J avian leukosis viruses
Genet. 4:e1000132. associated with hemangioma in Chinese layer hens.
275 Neiman, P.E, L. Jordan, R.A Weiss, and L.N Payne. Virol J. 8:552.
1980. Malignant lymphoma of the bursa of Fabricius: 288 Pandiri, A.R, I.M Gimeno, J.K Mays, W.M Reed, and
Analysis of early transformation. In: Cold Spring A.M Fadly. 2012. Reversion to subgroup J avian
Harbor Conference Cell Proliferation. Cold Spring leukosis virus viremia in seroconverted adult meat‐
Harbor Laboratory New York. 519–528. type chickens exposed to chronic stress by
276 Neiman, P.E, R. Kimmel, A. Icreverzi, K. Elsaesser, adrenocorticotrophin treatment. Avian Dis.
S.J Bowers, J. Burnside, and J. Delrow. 2006. 56:578–582.
Genomic instability during Myc‐induced 289 Pandiri, A.R, I.M Gimeno, W.M Reed, S.D Fitzgerald,
lymphomagenesis in the bursa of Fabricius. and A.M Fadly. 2009. Subgroup J avian leukosis virus‐
Oncogene. 25(47):6325–6335. induced histiocytic sarcomatosis occurs only in
277 Neumann, U., and R.L Witter. 1979. Differential persistently viremic but not immunotolerized
diagnosis of lymphoid leukosis and Marek’s disease by meat‐type chickens. Vet Pathol. 46:282–287.
tumor‐associated criteria. I. Studies on experimentally 290 Pandiri, A.R, J.K Mays, R.F Silva, H.D Hunt, W.M
infected chickens. Avian Dis. 23::417–425. Reed, and A.M Fadly. 2010. Subgroup J avian leukosis
278 Neumann, U., and R.L Witter. 1979. Differential virus neutralizing antibody escape variants contribute
diagnosis of lymphoid leukosis and Marek’s disease by to viral persistence in meat‐type chickens. Avian Dis.
tumor‐associated criteria. II. Studies on field cases. 54:848–856.
Avian Dis. 23:426–433. 291 Pandiri, A.R, W.M Reed, J.K Mays, and A.M Fadly.
279 Ning, Z.Y, Y.F An, W.B Qi, H. Wang, J.Q Pan, X.T Wu, 2007. Influence of strain, dose of virus, and age at
and M. Liao. 2012. Na(+)/H (+) exchanger 1 gene inoculation on subgroup J avian leukosis virus
expression in tissues of yellow chicken. Biochem persistence, antibody response, and oncogenicity in
Genet. 50:227–234. commercial meat‐type chickens. Avian Dis.
280 Nouvion, A.L, J. Thibaut, O.D Lohez, S. Venet, P. 51:725–732.
Colas, G. Gillet, and P. Lalle. 2007. Modulation of 292 Panet, A., D. Baltimore, and T. Hanafusa. 1975.
Nr‐13 antideath activity by peptide aptamers. Quantitation of avian RNA tumor virus reverse
Oncogene. 26:701–710. transcriptase by radioimmunoassay. J Virol.
281 Ochiai, K., K. Ohashi, T. Mukai, T. Kimura, T. 16:146–152.
Umemura, and C. Itakura. 1999. Evidence of 293 Pani, P.K, and P.M Biggs. 1973. Genetic control of
neoplastic nature and viral aetiology of so‐called fowl susceptibility to an A subgroup sarcoma virus in
glioma. Vet Rec. 145:79–81. commercial chickens. Avian Pathol. 2:27–41.
282 Okazaki, W., H.G Purchase, and B.R Burmester. 1975. 294 Payne, F.E, J.J Solomon, and H.G Purchase. 1966.
Phenotypic mixing test to detect and assay avian Immunofluorescent studies of group‐specific antigen
leukosis viruses. Avian Dis. 19:311–317. of the avian sarcoma‐leukosis viruses. Proc Natl Acad
283 Okazaki, W., H.G Purchase, and L.B Crittenden. 1982. Sci USA. 55:341–349.
Pathogenicity of avian leukosis viruses. Avian Dis. 295 Payne, L.N 1981. Immunity to lymphoid leukosis,
26:553–559. Rous sarcoma, and reticuloendotheliosis. In: Avian
Chapter 15  Neoplastic Diseases 695

Immunology. M.E Rose, L.N Payne, and B.M Freeman 310 Perek, M. 1960. An epizootic of histiocytic sarcomas
(eds.). British Poultry Science: Edinburgh, Scotland. in chickens induced by a cell‐free agent. Avian Dis.
285–299. 4:85–94.
296 Payne, L.N 1987. Epizootiology of avian leukosis virus 311 Perilla, J.R, and A.M Gronenborn. 2016. Molecular
infections. In: Avian Leukosis. G.F De Boer, ed. architecture of the retroviral capsid. Trends Biochem
Martinus Nijhoff, Boston. 47–76. Sci. 41:410–420.
297 Payne, L.N 1992. Biology of avian retroviruses. In: 312 Piraino, F., W. Okazaki, B.R Burmester, and T.N
The Retroviridae. J.A Levy, ed. Plenum Press, Fredrickson. 1963. Bioassay of Fowl leukosis virus in
New York. 299–404. chickens by the inoculation of 11‐day‐old embryos.
298 Payne, L.N 1992. Biology of avian retroviruses. In: Virology. 21:396–401.
The Retroviridae, vol 1. J. Levy (ed.). Plenum Press: 313 Pizer, E., and E.H Humphries. 1989. RAV‐1 insertional
New York. 299–404. mutagenesis: disruption of the c‐myb locus and
299 Payne, L.N, S.R Brown, N. Bumstead, K. Howes, J.A development of avian B‐cell lymphomas. J Virol.
Frazier, and M.E Thouless. 1991. A novel subgroup of 63:1630–1640.
exogenous avian leukosis virus in chickens. J Gen 314 Plachy, J., J. Kotab, P. Divina, M. Reinisova, F. Senigl,
Virol. 72(4):801–807. and J. Hejnar. 2010. Proviruses selected for high and
300 Payne, L.N, and N. Bumstead. 1982. Theoretical stable expression of transduced genes accumulate in
considerations on the relative importance of vertical broadly transcribed genome areas. J Virol. 84:4204–4211.
and horizontal transmission for the maintenance of 315 Plachy, J., M. Reinisova, D. Kucerova, F. Senigl, V.
infection by exogenous avian lymphoid leukosis virus. Stepanets, T. Hron, K. Trejbalova, D. Elleder, and J.
Avian Pathol. 11:547–553. Hejnar. 2017. Identification of New World Quails
301 Payne, L.N, A.M Gillespie, and K. Howes. 1992. susceptible to infection with avian leukosis virus
Myeloid leukaemogenicity and transmission of the subgroup J. J Virol. 91.
HPRS‐103 strain of avian leukosis virus. Leukemia. 316 Ponten, J. 1962. Transmission in vivo of chicken
6:1167–1176. erythroblastosis by intact cells. J Cell Comp Physiol.
302 Payne, L.N, A.M Gillespie, and K. Howes. 1993. 60:209–215.
Recovery of acutely transforming viruses from 317 Pope, C.R, E.M Odor, and M. Salem. 1999. Unusual
myeloid leukosis induced by the HPRS‐103 strain of eye lesions associated with ALV‐J virus. In:
avian leukosis virus. Avian Dis. 37:438–450. Proceedings, 28th Western Poultry Disease
303 Payne, L.N, A.M Gillespie, and K. Howes. 1993. Conference. 96–97.
Unsuitability of chicken sera for detection of 318 Powell, P.C, L.N Payne, J.A Frazier, and M. Rennie.
exogenous ALV by the group‐specific antigen ELISA. 1974. T lymphoblastoid cell lines from Marek’s disease
Vet Rec. 132:555–557. lymphomas. Nature. 251:79–80.
304 Payne, L.N, A.E Holmes, K. Howes, M. Pattison, D.L 319 Praharaj, N., C. Beaumont, G. Dambrine, D. Soubieux,
Pollock, and D.E Waters. 1982. Further studies on the L. Merat, D. Bouret, G. Luneau, J.M Alletru, M.H
eradication and epizootiology of lymphoid leukosis Pinard‐Van der Laan, P. Thoraval, and S. Mignon‐
virus infection in a commercial strain of chickens. Grasteau. 2004. Genetic analysis of the growth curve
Avian Pathol. 11:145–162. of Rous sarcoma virus‐induced tumors in chickens.
305 Payne, L.N, K. Howes, and D.F Adene. 1985. A Poult Sci. 83:1479–1488.
modified feather pulp culture method for determining 320 Price, J.A, and R.E Smith. 1981. Influence of
the genetic susceptibility of adult chickens to leukosis‐ bursectomy on bone growth and anemia induced by
sarcoma viruses. Avian Pathol. 14:261–265. avian osteopetrosis viruses. Cancer Res. 41:752–759.
306 Payne, L.N, and V. Nair. 2012. The long view: 40 years 321 Purchase, H.G 1987. Pathogenesis and pathology of
of avian leukosis research. Avian Pathol. 41:1–9. neoplasms caused by avian leukosis viruses. In: Avian
307 Payne, L.N, and M. Rennie. 1975. B cell antigen Leukosis. G.F De Boer, ed. Martinus Nijhoff, Boston.
markers on avian lymphoid leukosis tumour cells. Vet 171–196.
Rec. 96:454–456. 322 Purchase, H.G, and A.M Fadly. 1980. Leukosis and
308 Payne, L.N, and K. Venugopal. 2000. Neoplastic sarcomas. In: Isolation and Identification of Avian
diseases: Marek’s disease, avian leukosis and Pathogens. S.B Hitchner, C.H Domermuth, H.G
reticuloendotheliosis. Rev Sci Tech. 19:544–564. Purchase, and J.E Williams (eds.). American
309 Pecenka, V., P. Pajer, V. Karafiat, P. Kasparova, J. Association of Avian Patholologists: Kennett Square,
Dudlova, and M. Dvorak. 2017. HRAS, EGFR, MET, PA. 54–58.
and RON genes are recurrently activated by provirus 323 Purchase, H.G, and W. Okazaki. 1964. Morphology of
insertion in liver tumors induced by the retrovirus foci produced by standard preparation of Rous
myeloblastosis‐associated virus 2. J Virol. 91. sarcoma virus. J Natl Cancer Inst. 32:579–586.
696 Section II  Viral Diseases

324 Qin, A., L.F Lee, A. Fadly, H. Hunt, and Z. Cui. 2001. 336 Rous, P. 1911. A sarcoma of the fowl transmissible by
Development and characterization of monoclonal an agent separable from tumor cells. J Exp Med.
antibodies to subgroup J avian leukosis virus. Avian 13:397—411.
Dis. 45:938–945. 337 Rovigatti, V.G, and S.M Astrin. 1983. Avian endogenous
325 Qin, L., Y. Gao, W. Ni, M. Sun, Y. Wang, C. Yin, X. Qi, viral genes. Curr Top Microbiol Immunol. 103:1–21.
H. Gao, and X. Wang. 2013. Development and 338 Rubin, H. 2011. The early history of tumor virology:
application of real‐time PCR for detection of subgroup Rous, RIF, and RAV. Proc Natl Acad Sci USA.
J avian leukosis virus. J Clin Microbiol. 51:149–154. 108:14389–14396.
326 Qiu, Y., K. Qian, H. Shen, W. Jin, and A. Qin. 2011. 339 Rubin, H., A. Cornelius, and L. Fanshier. 1961. The
Development and validation of an indirect enzyme‐ pattern of congenital transmission of an avian leukosis
linked immunosorbent assay for the detection of avian virus. Proc Natl Acad Sci USA. 47:1058–1060.
leukosis virus antibodies based on a recombinant 340 Rubin, H., L. Fanshier, A. Cornelius, and W.F Huges.
capsid protein. J Vet Diagn Invest. 23:991–993. 1962. Tolerance and immunity in chickens after
327 Reinisova, M., J. Plachy, D. Kucerova, F. Senigl, M. congenital and contact infection with an avian
Vinkler, and J. Hejnar. Genetic 2016. Diversity of leukosis virus. 17:143—156. Virology. 17:143–156.
NHE1, receptor for subgroup J avian leukosis virus, in 341 Rup, B.J, J.D Hoelzer, and H.R Bose, Jr. 1982. Helper
domestic chicken and wild anseriform species. PLoS viruses associated with avian acute leukemia viruses
One. 11:e0150589. inhibit the cellular immune response. Virology.
328 Reinisova, M., J. Plachy, K. Trejbalova, F. Senigl, D. 116:61–71.
Kucerova, J. Geryk, J. Svoboda, and J. Hejnar. 2012. 342 Rutherford, K., and B.F Benkel. 2013. Characterization
Intronic deletions that disrupt mRNA splicing of the of insertion sites and development of locus‐specific
tva receptor gene result in decreased susceptibility to assays for three broiler‐derived subgroup E avian
infection by avian sarcoma and leukosis virus leukosis virus proviruses. Avian Pathol. 42:373–378.
subgroup A. J Virol. 86:2021–2030. 343 Rutherford, K., N. McLean, and B.F Benkel. 2014. A
329 Reinisova, M., F. Senigl, X. Yin, J. Plachy, J. Geryk, D. rapid profiling assay for avian leukosis virus subgroup
Elleder, J. Svoboda, M.J Federspiel, and J. Hejnar. 2008. E proviruses in chickens. Avian Dis. 58:34–38.
A single‐amino‐acid substitution in the TvbS1 344 Rutherford, K., C.J Meehan, M.G Langille, S.G Tyack,
receptor results in decreased susceptibility to J.C McKay, N.L McLean, K. Benkel, R.G Beiko, and B.
infection by avian sarcoma and leukosis virus Benkel. 2016. Discovery of an expanded set of avian
subgroups B and D and resistance to infection by leukosis subroup E proviruses in chickens using
subgroup E in vitro and in vivo. J Virol. 82:2097–2105. Vermillion, a novel sequence capture and analysis
330 Resnick‐Roguel, N., H. Burstein, J. Hamburger, A. pipeline. Poult Sci. 95:2250–2258.
Panet, A. Eldor, I. Vlodavsky, and M. Kotler. 1989. 345 Sacco, M.A, K. Howes, L.P Smith, and V.K Nair. 2004.
Cytocidal effect caused by the envelope glycoprotein Assessing the roles of endogenous retrovirus EAV‐HP
of a newly isolated avian hemangioma‐inducing in avian leukosis virus subgroup J emergence and
retrovirus. J Virol. 63:4325–4330. tolerance. J Virol. 78:10525–10535.
331 Resnick‐Roguel, N., A. Eldor, H. Burstein, E. Hy‐Am, 346 Sacco, M.A, and V.K Nair. 2014. Prototype
I. Vlodavsky, A. Panet, M.A Blajchman, and M. Kotler. endogenous avian retroviruses of the genus Gallus. J
1990. Envelope glycoprotein of avian hemangioma Gen Virol. 95:2060–2070.
retrovirus induces a thrombogenic surface on human 347 Salter, D.W, A. Fadly, E. Smith, K. Nazerian, N.
and bovine endothelial cells. J Virol. 64:4029–4032. Yanagida, R. Silva, D. Reilly, D. Marshall, L. Bacon,
332 Robertson, J.S, C. Nicolson, A.M Riley, M. Bentley, G. and L. Crittenden. 1991. The use of vaccines and
Dunn, T. Corcoran, G.C Schild, and P. Minor. 1997. genetic resistance (natural and transgenic) to control
Assessing the significance of reverse transcriptase avian leukosis. In D. Swayne and D. Zander (eds.).
activity in chick cell‐derived vaccines. Biologicals. Proceedings Avian Tumor Virus Symposium. 9–14.
25:403–414. 348 Sandelin, K., and T. Estola. 1974. Occurrence of
333 Robinson, H.L, L. Ramamoorthy, K. Collart, and D.W different subgroups of avian leukosis virus in Finnish
Brown. 1993. Tissue tropism of avian leukosis viruses: poultry. Avian Pathol. 3:159—168.
analyses for viral DNA and proteins. Virology. 193:443–445. 349 Sanger, V.L, T.N Fredrickson, C.C Morrill, and B.R
334 Roloff, F. Mag Ges 1868. Thierheilkd 34:190 (cited by Burmester. 1966. Pathogenesis of osteopetrosis in
Chubb, L.G, and R.F Gordon. 1957). Vet Rev Annot. chickens. Am J Vet Res. 27:1735–1744.
32:97–120. 350 Schat, K.A 1987. Immunity in Marek’s disease and
335 Roth, F.K, P. Meyers, and R.M Dougherty. 1971. The other tumors. In: Avian Immunology: Basis and
presence of avian leukosis virus group‐specific Practice. A. Toivanen and P. Toivanen (eds.). CRC
antibodies in chicken sera. Virology. 45:265–274. Press: Boca Raton, FL Vol. II. 101–128.
Chapter 15  Neoplastic Diseases 697

351 Schat, K.A 1996. Immunity to Marek’s disease, 364 Smith, E.J, A. Fadly, and W. Okazaki. 1979. An
lymphoid leukosis and reticuloendotheliosis. In: enzyme‐linked immunosorbent assay for detecting
Poultry Immunology. T.F Davison, T.R Morris, and avian leukosis‐sarcoma viruses. Avian Dis.
L.N Payne (eds.). Carfax Publishing Company: 23:698–707.
Abingdon. 209–233. 365 Smith, E.J, and A.M Fadly. 1988. Influence of
352 Schat, K.A, and H.N Erb. 2014. Lack of evidence that congenital transmission of endogenous virus‐21 on
avian oncogenic viruses are infectious for humans: a the immune response to avian leukosis virus infection
review. Avian Dis. 58:345–358. and the incidence of tumors in chickens. Poult Sci.
353 Schierman, L.W, and W.M Collins. 1987. Influence of 67:1674–1679.
the major histocompatibility complex on tumor 366 Smith, E.J, and A.M Fadly. 1994. Male‐mediated
regression and immunity in chickens. Poult Sci. venereal transmission of endogenous avian leukosis
66:812–818. virus. Poult Sci. 73:488–494.
354 Schmidt, E.V, and R.E Smith. 1981. Avian 367 Smith, E.J, A.M Fadly, and L.B Crittenden. 1986.
osteopetrosis virus induces proliferation of cultured Observations on an enzyme‐linked immunosorbent
bone cells. Virology. 111:275–282. assay for the detection of antibodies against avian
355 Schulten, E.S, W.E Briles, and R.L Taylor, Jr. 2009. leukosis‐sarcoma viruses. Avian Dis. 30:488–493.
Rous sarcoma growth in lines congenic for major 368 Smith, E.J, U. Neumann, and W. Okazaki. 1980.
histocompatibility (B) complex recombinants. Poult Immune response to avian leukosis virus infection in
Sci. 88:1601–1607. chickens: Sequential expression of serum
356 Schur, F.K, R.A Dick, W.J Hagen, V.M Vogt, and J.A immunoglobulins and viral antibodies. Comp
Briggs. 2015. The structure of immature virus‐like Immunol Microbiol Infect Dis. 2:519–529.
Rous sarcoma virus gag particles reveals a structural 369 Smith, E.J, S.M Williams, and A.M Fadly. 1998.
role for the p10 domain in assembly. J Virol. Detection of avian leukosis virus subgroup J using the
89:10294–10302. polymerase chain reaction. Avian Dis. 42:375–380.
357 Segura, J.C, J.S Gavora, J.L Spencer, R.W Fairfull, R.S 370 Smith, L.M, S.R Brown, K. Howes, S. McLeod, S.S
Gowe, and R.B Buckland. 1988. Semen traits and Arshad, G.S Barron, K. Venugopal, J.C McKay, and
fertility of White Leghorn males shown to be positive L.N Payne. 1998. Development and application of
or negative for lymphoid leukosis virus in semen and polymerase chain reaction (PCR) tests for the
feather pulp. Br Poult Sci. 29:545–553. detection of subgroup J avian leukosis virus. Virus Res.
358 Shank, P.R, P.J Schatz, L.M Jensen, P.N Tsichlis, J.M 54:87–98.
Coffin, and H.L Robinson. 1985. Sequences in the 371 Smith, R.E 1987. Immunology of avian leukosis virus
gag‐pol‐5’env region of avian leukosis viruses confer infections. In: Avian Leukosis. G.F De Boer, ed.
the ability to induce osteopetrosis. Virology. Martinus Nijhoff, Boston. 121–130.
145:94–104. 372 Soffer, D., N. Resnick‐Roguel, A. Eldor, and M. Kotler.
359 Sigel, M.M, P. Meyers, and H.T Holden. 1971. 1990. Multifocal vascular tumors in fowl induced by a
Resistance to Rous sarcoma elicited by immunization newly isolated retrovirus. Cancer Res. 50:4787–4793.
with live virus. Proc Soc Exp Biol Med. 137:142–146. 373 Spencer, J.L 1984. Progress towards eradication of
360 Silva, R.F, A.M Fadly, and H.D Hunt. 2000. lymphoid leukosis viruses—a review. Avian Pathol.
Hypervariability in the envelope genes of subgroup J 13:599–619.
avian leukosis viruses obtained from different farms in 374 Spencer, J.L, M. Chan, and S. Nandin‐Davis. 2000.
the United States. Virology. 272:106–111. Relationship between egg size and subgroup J avian
361 Silva, R.F, A.M Fadly, and S.P Taylor. 2007. leukosis virus in eggs from broiler breeders. Avian
Development of a polymerase chain reaction to Pathol. 29:617–622.
differentiate avian leukosis virus (ALV) subgroups: 375 Spencer, J.L, L.B Crittenden, B.R Burmester, W.
detection of an ALV contaminant in commercial Okazaki, and R.L Witter. 1977. Lymphoid leukosis:
Marek’s disease vaccines. Avian Dis. 51:663–667. interrelations among virus infections in hens, eggs,
362 Simon, M.C, W.S Neckameyer, W.S Hayward, and R.E embryos, and chicks. Avian Dis. 21:331–345.
Smith. 1987. Genetic determinants of neoplastic 376 Spencer, J.L, J.S Gavora, and R.S Gowe. 1980.
diseases induced by a subgroup F avian leukosis virus. Lymphoid leukosis virus: natural transmission and
J Virol. 61:1203–1212. nonneoplastic effects. In: Cold Spring Harbor
363 Smith, E.J, and L.B Crittenden. 1988. Genetic cellular Conference on Cell Proliferation. Cold Spring Harbor,
resistance to subgroup E avian leukosis virus in slow‐ New York. 553–564.
feathering dams reduces congenital transmission of an 377 Stedman, N.L, and T.P Brown. 1999. Body weight
endogenous retrovirus encoded at locus ev21. Poult suppression in broilers naturally infected with avian
Sci. 67:1668–1673. leukosis virus subgroup J. Avian Dis. 43:604–610.
698 Section II  Viral Diseases

378 Stedman, N.L, and T.P Brown. 2002. Cardiomyopathy 391 Svoboda, J. 2016. On board a raft or boat in the
in broiler chickens congenitally infected with avian retrovirus sea. Proc Natl Acad Sci USA.
leukosis virus subgroup J. Vet Pathol. 39:161–164. 113:3927–3931.
379 Stedman, N.L, T.P Brown, and D.I Bounous. 2000. 392 Svoboda, J., J. Hejnar, J. Geryk, D. Elleder, and Z.
Functions of heterophils, macrophages, and Vernerova. 2000. Retroviruses in foreign species and
lymphocytes isolated from broilers naturally infected the problem of provirus silencing. Gene. 261:181–188.
with avian leukosis virus subgroup J. In: E.F Kaleta, 393 Swanstrom, R., W.D Graham, and S. Zhou. 2017.
L.N Payne, and U. Heffels‐Redmann (eds.). Sequencing the Biology of entry: the retroviral env
Proceedings, International Symposium on ALV‐J and gene. Curr Top Microbiol Immunol. 407:65.
Other Avian Retroviruses. Rauischholzhausen, 394 Swanstrom, R., and J.W Wills. 1997. Synthesis,
Germany. 111–114. assembly and processing of viral proteins. In:
380 Stedman, N.L, T.P Brown, R.L Brooks, Jr., and D.I Retroviruses. J.M Coffin, S.H Hughes and H.E Varmus,
Bounous. 2001. Heterophil function and resistance to eds. Cold Spring Harbor, New York. 263–334.
staphylococcal challenge in broiler chickens naturally 395 Tam, W., D. Ben‐Yehuda, and W.S 1997. Hayward. bic,
infected with avian leukosis virus subgroup J. Vet a novel gene activated by proviral insertions in avian
Pathol. 38:519–527. leukosis virus‐induced lymphomas, is likely to
381 Stedman, N.L, T.P Brown, and C.C Brown. 2001. function through its noncoding RNA. Mol Cell Biol.
Localization of avian leukosis virus subgroup J in 17:1490–1502.
naturally infected chickens by RNA in situ 396 Tam, W., and J.E Dahlberg. 2006. miR‐155/BIC as an
hybridization. Vet Pathol. 38:649–656. oncogenic microRNA. Genes Chromosomes Cancer.
382 Stepanets, V., Z. Vernerova, M. Vilhelmova, J. Geryk, 45:211–212.
J. Plachy, J. Hejnar, F.F Weichold, and J. Svoboda. 397 Tam, W., S.H Hughes, W.S Hayward, and P. Besmer.
2003. Intraembryonic avian leukosis virus subgroup C 2002. Avian bic, a gene isolated from a common
(ALV‐C) inoculation producing wasting disease in retroviral site in avian leukosis virus‐induced
ducks soon after hatching. Folia Biol (Praha). 49:100–109. lymphomas that encodes a noncoding RNA,
383 Sultana, T., A. Zamborlini, G. Cristofari, and P. cooperates with c‐myc in lymphomagenesis and
Lesage. 2017. Integration site selection by retroviruses erythroleukemogenesis. J Virol. 76:4275–4286.
and transposable elements in eukaryotes. Nature Rev 398 Taylor, R.L, Jr. 2004. Major histocompatibility (B)
Genet. 18:292–308. complex control of responses against Rous sarcomas.
384 Sun, M., D. Yu, H. Mo, H. Cao, C. Chen, and F. Chen. Poult Sci. 83:638–649.
Epitope 2012. Mapping of a monoclonal antibody 399 Temin, H.M 1974. The cellular and molecular biology
against the gp85 of avian leukosis virus subgroup J. of RNA tumor viruses, especially avian leukosis‐
J Vet Med Sci. 74(6):693–697. sarcoma viruses, and their relatives. Adv Cancer Res.
385 Sun, Y.H, L.H Xie, X. Zhuo, Q. Chen, D. Ghoneim, B. 19:47–104.
Zhang, J. Jagne, C. Yang, and X.Z Li. 2017. Domestic 400 Temin, H.M, and H. Rubin. 1958. Characteristics of an
chickens activate a piRNA defense against avian assay for Rous sarcoma virus and Rous sarcoma cells
leukosis virus. eLife 6. in tissue culture. Virology. 6:669–688.
386 Suzuki, K., E. Kobayashi, H. Yamashita, H. Uenishi, I. 401 Tereba, A., and K.G Murti. 1977. A very sensitive
Churkina, G. Plastow, N. Hamasima, and T. biochemical assay for detecting and quantitating avian
Mitsuhashi. 2012. Structural analysis of MHC alleles oncornaviruses. Virology. 80:166–176.
in an RSV tumour regression chicken using a BAC 402 Toyoda, T., K. Ochiai, H. Hatai, M. Murakami, E. Ono,
library. Anim Genet. 43:348–351. T. Kimura, and T. Umemura. 2006. Cerebellar
387 Suzuki, K., T. Matsumoto, E. Kobayashi, H. Uenishi, I. hypoplasia associated with an avian leukosis virus
Churkina, G. Plastow, H. Yamashita, N. Hamasima, inducing fowl glioma. Vet Pathol. 43:294–301.
and T. Mitsuhashi. 2010. Genotypes of chicken major 403 Trejbalova, K., K. Gebhard, Z. Vernerova, L. Dusek, J.
histocompatibility complex B locus associated with Geryk, J. Hejnar, A.T Haase, and J. Svoboda. 1999.
regression of Rous sarcoma virus J‐strain tumors. Proviral load and expression of avian leukosis viruses
Poult Sci. 89:651–657. of subgroup C in long‐term persistently infected
388 Svoboda, J. 2008. Foundations in cancer research. The heterologous hosts (ducks). Arch Virol.
turns of life and science. Adv Cancer Res. 99:1–32. 144:1779–1807.
389 Svoboda, J. 2013. Rous sarcoma virus centennial in 404 Tsang, S.X, W.M Switzer, V. Shanmugam, J.A Johnson,
Folia Biologica. Folia Biol (Praha). 59:103–104. C. Goldsmith, A. Wright, A. Fadly, D. Thea, H. Jaffe,
390 Svoboda, J. Cell. 2015. Association in Rous sarcoma T.M Folks, and W. Heneine. 1999. Evidence of avian
virus (RSV) rescue and cell infection. Folia Biol leukosis virus subgroup E and endogenous avian virus
(Praha). 61:161–167. in measles and mumps vaccines derived from chicken
Chapter 15  Neoplastic Diseases 699

cells: investigation of transmission to vaccine 418 Wallny, H.J, D. Avila, L.G Hunt, T.J Powell, P. Riegert,
recipients. J Virol. 73:5843–5851. J. Salomonsen, K. Skjodt, O. Vainio, F. Vilbois, M.V
405 Tsukamoto, K., M. Hasebe, S. Kakita, H. Hihara, and Wiles, and J. Kaufman. 2006. Peptide motifs of the
Y. Kono. 1991. Identification and characterization of single dominantly expressed class I molecule explain
hens transmitting avian leukosis virus (ALV) to their the striking MHC‐determined response to Rous
embryos by ELISAs for detecting infectious ALV, ALV sarcoma virus in chickens. Proc Natl Acad Sci USA.
antigens and antibodies to ALV. J Vet Med Sci. 103:1434–1439.
53:859–864. 419 Walter, W.G, B.R Burmester, and C.H Cunningham.
406 Tsukamoto, K., M. Hasebe, S. Kakita, Y. Taniguchi, H. 1962. Studies on the transmission and pathology of a
Hihara, and Y. Kono. 1992. Sporadic congenital viral‐induced avian nephroblastoma (embryonal
transmission of avian leukosis virus in hens nephroma). Avian Dis. 6:455–477.
discharging the virus into the oviducts. J Vet Med Sci. 420 Wang, F., X. Wang, H. Chen, J. Liu, and Z. Cheng.
54:99–103. 2011. The critical time of avian leukosis virus
407 Tsukamoto, K., Y. Kono, and K. Arai. 1985. An subgroup J‐mediated immunosuppression during
enzyme linked immunosorbent assay for detection of early stage infection in specific pathogen‐free
antibodies to exogenous avian leukosis virus. Avian chickens. J Vet Sci. 12:235–241.
Dis. 29:1118–1129. 421 Wang, G., Y. Jiang, L. Yu, Y. Wang, X. Zhao, and Z.
408 Van Woensel, P.A M., A. v. Blaaderen, R.J M. Cheng. 2013. Avian leukosis virus subgroup J
Mooman, and G.F d. Boer. 1992. Detection of proviral associated with the outbreak of erythroblastosis in
DNA and viral RNA in various tissues early after avian chickens in China. Virol J. 10:92.
leukosis virus infection. Leukemia. 6(3). 422 Wang, Y., L. Fang, J. Li, Y. Li, S. Cui, X. Sun, S. Chang,
409 Varmus, H. 2017. How Tumor virology evolved into P. Zhao, and Z. Cui. 2016. Rescue of avian leukosis
cancer biology and transformed oncology. Ann Rev subgroup‐J‐associated acutely transforming viruses
Cancer Biol. 1:1–18. carrying different lengths of the v‐fps oncogene and
410 Venugopal, K., K. Howes, G.S Barron, and L.N Payne. analysis of their tumorigenicity. Arch Virol.
1997. Recombinant env‐gp85 of HPRS‐103 (subgroup 161:3473–3481.
J) avian leukosis virus: antigenic characteristics and 423 Wang, Y., Z. Kang, Y. Gao, L. Qin, L. Chen, Q. Wang,
usefulness as a diagnostic reagent. Avian Dis. J. Li, H. Gao, X. Qi, H. Lin, and X. Wang. 2011.
41:283–288. Development of loop‐mediated isothermal
411 Venugopal, K., K. Howes, D.M J. Flannery, and L.N amplification for rapid detection of avian leukosis
Payne. 2000. Isolation of acutely transforming virus subgroup A. J Virol Methods. 173:31–36.
subgroup J avian leukosis viruses that induce 424 Wang, Y., J. Li, Y. Li, L. Fang, X. Sun, S. Chang, P.
erythroblastosis and myelocytomatosis. Avian Pathol. Zhao, and Z. Cui. 2016. Identification of avian leukosis
29:327–332. virus subgroup J‐associated acutely transforming
412 Venugopal, K., K. Howes, D.M J. Flannery, and L.N viruses carrying the v‐src oncogene in layer chickens.
Payne. Subgroup 2000. J avian leukosis virus infection J Gen Virol. 97:1240–1248.
in turkeys: induction of rapid onset tumours by 425 Wang, Y., Y. Yin, X. Lan, F. Ye, K. Tian, X. Zhao, H.
acutely transforming virus strain 966. Avian Pathol. Yin, D. Li, H. Xu, Y. Liu, and Q. Zhu. 2017. Molecular
29:319–326. characterization, expression of chicken TBK1 gene
413 Victoria, J.G, C. Wang, M.S Jones, C. Jaing, K. and its effect on IRF3 signaling pathway. PLoS One.
McLoughlin, S. Gardner, and E.L Delwart. 2010. Viral 12:e0177608.
nucleic acids in live‐attenuated vaccines: detection of 426 Watts, S.L, and R.E Smith. 1980. Pathology of
minority variants and an adventitious virus. J Virol. chickens infected with avian nephoblastoma virus
84:6033–6040. MAV‐2(N). Infect Immun. 27:501–512.
414 Vogt, P.K 1965. Avian tumor viruses. Adv Virus Res. 427 Weiss, R.A, and P.K 2011. Vogt. 100 years of Rous
11:293–385. sarcoma virus. J Exp Med. 208:2351–2355.
415 Vogt, P.K 2010. Oncogenes and the revolution in 428 Weissmahr, R.N, J. Schupbach, and J. Boni. 1997.
cancer research: homage to hidesaburo hanafusa Reverse transcriptase activity in chicken embryo
(1929–2009). Genes Cancer. 1:6–11. fibroblast culture supernatants is associated with
416 Vogt, P.K, A.G Bader, and S. Kang. 2006. particles containing endogenous avian retrovirus
Phosphoinositide 3‐kinase: from viral oncoprotein to EAV‐0 RNA. J Virol. 71:3005–3012.
drug target. Virology. 344:131–138. 429 Whalen, L.R, D.W Wheeler, D.H Gould, S.A Fiscus,
417 Vogt, V.M 2017. Retroviral Virions and Genomes. Cold L.C Boggie, and R.E Smith. 1988. Functional and
Spring Harbor Laboratory Press, Cold Spring Harbor, structural alterations of the nervous system induced by
NY. avian retrovirus RAV‐7. Microbial Pathog. 4:401–416.
700 Section II  Viral Diseases

430 Wilczek, C., O. Chayka, A. Plachetka, and K.H American Association of Avian Pathologists, Athens,
Klempnauer. 2009. Myb‐induced chromatin GA (Electronic media). 1–49.
remodeling at a dual enhancer/promoter element 442 Wright, S.E, and D.D Bennett. 1992. Avian retroviral
involves non‐coding rna transcription and is recombinant expressing foreign envelope delays
disrupted by oncogenic mutations of v‐myb. J Biol tumour formation of ASV‐A‐induced sarcoma.
Chem. 284:35314–35324. Vaccine. 10:375–378.
431 Williams, S.M, T. Barbosa, S. Hafner, and G. Zavala. 443 Wu, X., K. Qian, A. Qin, H. Shen, P. Wang, W. Jin, and
2010. Myxosarcomas associated with avian leukosis Y.M Eltahir. 2010. Recombinant avian leukosis viruses
virus subgroup A infection in fancy breed chickens. of subgroup J isolated from field infected commercial
Avian Dis. 54:1319–1322. layer chickens with hemangioma and myeloid leukosis
432 Williams, S.M, S.D Fitzgerald, W.M Reed, L.F Lee, and possess an insertion in the E element. Vet Res
A.M Fadly. 2004. Tissue tropism and bursal Commun. 34:619–632.
transformation ability of subgroup J avian leukosis 444 Xie, Q., J. Zhang, H. Shao, Z. Wan, X. Tian, J. Yang, M.
virus in White Leghorn chickens. Avian Dis. Pang, K. Qian, W. Gao, C. Wang, A. Qin, and J. Ye.
48:921–927. 2016. Development of a novel immuno‐PCR for
433 Williams, S.M, W.M Reed, L.D Bacon, and A.M Fadly. detection of avian leukosis virus. J Virol Methods.
2004. Response of white leghorn chickens of various 236:25–28.
genetic lines to infection with avian leukosis virus 445 Yang, F., R.R Xian, Y. Li, T.S Polony, and K.L Beemon.
subgroup J. Avian Dis. 48:61–67. 2007. Telomerase reverse transcriptase expression
434 Williams, S.M, W.M Reed, and A.M Fadly 2000. elevated by avian leukosis virus integration in B cell
Influence of age of exposure on the response of line 0 lymphomas. Proc Natl Acad Sci USA.
and line 63 chickens to infection with subgroup J 104:18952–18957.
avian leukosis virus. In: E.F Kaleta, L.N Payne, and U. 446 Zander, D.V, R.G Raymond, C.F McClary, and K.
Heffels‐Redmann (eds.). Proceedings, International Goodwin. 1975. Eradication of subgroups A and B
Symposium on ALV‐J and Other Avian Retroviruses; lymphoid leukosis virus from commercial poultry
Rauischholzhausen, Germany:67–76. breeding flocks. Avian Dis. 19:403–423.
435 Winans, S., R.C Larue, C.M Abraham, N. Shkriabai, 447 Zavala, G., S. Cheng, and T. Barbosa. 2012. Natural
A. Skopp, D. Winkler, M. Kvaratskhelia, and K.L infection and transmission of a retrovirus closely
Beemon. 2017. The FACT complex promotes avian related to myeloblastosis‐associated virus type 1 in
leukosis virus DNA integration. J Virol. 91. egg‐type chickens. Avian Dis. 56:7–14.
436 Witter, R.L 2000. Determinants of early transmission 448 Zavala, G., B. Lucio‐Martinez, S. Cheng, and T.
of ALV‐J in commercial broiler breeder chickens. In: Barbosa. 2006. Sarcomas and myelocytomas induced
Proceedings, International, Symposiumon ALV‐J and by a retrovirus related to myeloblastosis‐associated
Other Avian Retroviruses. E.F Kaleta, L.N Payne and virus type 1 in White Leghorn egg layer chickens.
U. Heffels‐Redmann, eds., Rauischholzhausen, Avian Dis. 50:201–208.
Germany. 216–225. 449 Zhang, D., H. Li, Z. Zhang, S. Sun, Z. Cheng, J. Liu, P.
437 Witter, R.L, L.D Bacon, H.D Hunt, R.E Silva, and A.M Zhao, Q. Ren, and H. Guo. 2015. Antibody responses
Fadly. 2000. Avian leukosis virus subgroup J infection induced by recombinant ALV‐A gp85 protein vaccine
profiles in broiler breeder chickens: association with combining with CpG‐ODN adjuvant in breeder hens
virus transmission to progeny. Avian Dis. 44:913–931. and the protection for their offspring against early
438 Witter, R.L, L.D Bacon, H.D Hunt, R.E Silva, and A.M infection. Antiviral Res. 116:20–26.
Fadly. 2001. Avian leukosis virus subgroup J infection 450 Zhang, H.M, L.D Bacon, H.H Cheng, and H.D Hunt.
profiles in broiler breeder chickens: Associations with 2005. Development and validation of a PCR‐RFLP
virus transmission to progeny. Avian Dis. 44:913–931. assay to evaluate TVB haplotypes coding receptors for
439 Witter, R.L, B.W Calnek, and P.P Levine. 1966. subgroup B and subgroup E avian leukosis viruses in
Influence of naturally occurring parental antibody on White Leghorns. Avian Pathol. 34:324–331.
visceral lymphomatosis virus infection in chickens. 451 Zhang, H.N, H.Z Lai, Y. Qi, X.T Zhang, Z.Y Ning,
Avian Dis. 10:43–56. K.J Luo, C.A Xin, W.S Cao, and M. Liao. 2011. An
440 Witter, R.L, and A.M Fadly. 2001. Reduction of ALV‐J isolate is responsible for spontaneous
horizontal transmission of avian leukosis virus haemangiomas in layer chickens in China. Avian
subgroup J in broiler breeder chickens hatched and Pathol. 40:261–267.
reared in small groups. Avian Pathol. 30:641–654. 452 Zhang, L., D. Cai, X. Zhao, Z. Cheng, H. Guo, C. Qi, J.
441 Witter, R.L, I.M Gimeno, and A.M Fadly 2005. Liu, R. Xu, P. Zhao, and Z. Cui. 2014. Liposomes
Differential diagnosis of lymphoid and myeloid containing recombinant gp85 protein vaccine against
tumors in the chicken. In: AAAP Slide Study Set #27 ALV‐J in chickens. Vaccine. 32:2452–2456.
Chapter 15  Neoplastic Diseases 701

453 Zhang, X., M. Liao, P. Jiao, K. Luo, H. Zhang, T. Ren, 10 Bagust, T.J, and T.M Grimes. 1979. Experimental
G. Zhang, C. Xu, C. Xin, and W. Cao. 2010. infection of chickens with an Australian strain of
Development of a loop‐mediated isothermal reticuloendotheliosis virus. 2. Serological responses
amplification assay for rapid detection of subgroup J and pathogenesis. Avian Pathol. 8:375–389.
avian leukosis virus. J Clin Microbiol. 48:2116–2121. 11 Bagust, T.J, T.M Grimes, and D.P Dennett. 1979.
54 Zhou, G., W. Cai, X. Liu, C. Niu, C. Gao, C. Si, W.
4 Infection studies on a reticuloendotheliosis virus
Zhang, L. Qu, and L. Han. 2011. A duplex real‐time contaminant of a commercial Marek’s disease vaccine.
reverse transcription polymerase chain reaction for Aust Vet J. 55:153–157.
the detection and quantitation of avian leukosis virus 12 Bagust, T.J, T.M Grimes, and N. Ratnamohan. 1981.
subgroups A and B. J Virol. Methods 173:275–279. Experimental infection of chickens with an Australian
strain of reticuloendotheliosis virus 3. Persistent
infection and transmission by the adult hen. Avian
Reticuloendotheliosis
Pathol. 10:375–385.
1 Alger, K., E. Bunting, K. Schuler, and C.M Whipps. 13 Bagust, T.J, J.B McFerran, and M.S McNulty. 1993.
2017. Risk factors for and spatial distribution of Virus infections of birds. In: Virus Infections of
lymphoproliferative disease virus (Lpdv) in wild turkeys Vertebrates. Elsevier, Amsterdam. 437–454.
(Meleagris Gallopavo) in New York State, USA. J Wildl 14 Bao, K.Y, Y.P Zhang, H.W Zheng, H.C Lv, Y.L Gao, J.F
Dis. 53:499–508. Wang, H.L Gao, X.L Qi, H.Y Cui, Y.Q Wang, X.G Ren,
2 Allen, P.T, J.A Mullins, C.L Harris, A. Hellman, R.F X.M Wang, and C.J Liu. 2015. Isolation and full‐
Garry, and M.R F. Waite. 1979. Replication of genome sequence of two reticuloendotheliosis virus
reticuloendotheliosis virus in mammalian cells strains from mixed infections with Marek’s disease
(abstract). In: Amer Soc Microbiol Annual Meeting. 256. virus in China. Virus Genes. 50:418–424.
3 Allison, A.B, M. Kevin Keel, J.E Philips, A.N Cartoceti, 15 Barbacid, M., E. Hunter, and S.A Aaronson. 1979. Avian
B.A Munk, N.M Nemeth, T.I Welsh, J.M Thomas, J.M reticuloendotheliosis viruses: evolutionary linkage with
Crum, A.B Lichtenwalner, A.M Fadly, G. Zavala, E.C mammalian type C retroviruses. J Virol. 30:508–514.
Holmes, and J.D Brown. 2014. Avian oncogenesis 16 Barbosa, T., G. Zavala, S. Cheng, T. Lourenco, and P.
induced by lymphoproliferative disease virus: a Villegas. 2006. Effects of reticuloendotheiolsis virus on
neglected or emerging retroviral pathogen? Virology. the viability and reproductive performance of Japanese
450–451:2–12. quail. J App Poult Res. 15:558–563.
4 Aly, M.M, M.K Hassan, A.A Elzahr, A.A Amin, and F.E 17 Barbosa, T., G. Zavala, S. Cheng, and P. Villegas. 2007.
Saad. 1998. Serological survey on reticuloendotheliosis Full genome sequence and some biological properties
virus infection in commercial chicken and turkey flocks of reticuloendotheliosis virus strain APC‐566 isolated
in Egypt. In: Proc. 5th Sci. Conference, Egypt Vet from endangered Attwater’s prairie chickens. Virus Res.
Poultry Association. Cairo, Egypt. 51–68. 124:68–77.
5 Aly, M.M, E.J Smith, and A.M Fadly. 1993. Detection of 18 Barbosa, T., G. Zavala, S. Cheng, and P. Villegas. 2007.
reticuloendotheliosis virus infection using the Pathogenicity and transmission of reticuloendotheliosis
polymerase chain reaction. Avian Pathol. 22:543–554. virus isolated from endangered prairie chickens. Avian
6 Aly, M.M, R.L Witter, and A.M Fadly. 1996. Dis. 51:33–39.
Enhancement of reticuloendotheliosis virus‐induced 19 Barth, C.F, D.L Ewert, W.C Olson, and E.H Humphries.
bursal lymphomas by serotype 2 Marek’s disease virus. 1990. Reticuloendotheliosis virus REV‐T(REV‐A)‐
Avian Pathol. 25:81–94. induced neoplasia: development of tumors within the
7 Aulisio, C.G, and A. Shelokov. 1969. Prevalence of T‐lymphoid and myeloid lineages. J Virol. 64:6054–6062.
reticuloendotheliosis in chickens: immunofluorescence 20 Barth, C.F, and E.H Humphries. 1988. Expression of
studies. Proc Soc Exp Biol Med. 130:178–181. v‐rel induces mature B‐cell lines that reflect the
8 Bacon, L.D, R.L Witter, and A.M Fadly. 1989. diversity of avian immunoglobulin heavy‐ and light‐
Augmentation of retrovirus‐induced lymphoid leukosis chain rearrangements. Mol Cell Biol. 8:5358–5368.
by Marek’s disease herpesviruses in White Leghorn 21 Barth, C.F, and E.H Humphries. 1988. A
chickens. J Virol. 63:504–512. nonimmunosuppressive helper virus allows high
9 Bader, S.R, S. Kothlow, S. Trapp, S.C Schwarz, H.C efficiency induction of B cell lymphomas by
Philipp, S. Weigend, A.R Sharifi, R. Preisinger, W. reticuloendotheliosis virus strain T. J Exp Med.
Schmahl, B. Kaspers, and K. Matiasek. 2010. Acute 167:89–108.
paretic syndrome in juvenile White Leghorn chickens 22 Bauer, G., and H.M Temin. 1980. Specific antigenic
resembles late stages of acute inflammatory relationships between the RNA‐dependent DNA
demyelinating polyneuropathies in humans. polymerases of avian reticuloendotheliosis viruses and
J Neuroinflammation. 7:7. mammalian type C retroviruses. J Virol. 34:168–177.
702 Section II  Viral Diseases

23 Baxter‐Gabbard, K.L, W.F Campbell, F. Padgett, A. envelope glycoprotein of an avian reticuloendotheliosis


Raitano‐Fenton, and A.S Levin. 1971. Avian retrovirus induce neutralizing antibodies and reduce
reticuloendotheliosis virus (strain T). II. Biochemical viremia in chickens. J Virol. 67:3069–3076.
and biophysical properties. Avian Dis. 15:850–862. 37 Campbell, W.F, K.L Baxter‐Gabbard, and A.S Levine.
24 Baxter‐Gabbard, K.L, D.A Peterson, A.S Levine, P. 1971. Avian reticuloendotheliosis virus (strain T). I.
Meyers, and M.M Sigel. 1973. Reticuloendotheliosis Virological characterization. Avian Dis. 15:837–849.
virus (strain T). VI. An immunogen versus 38 Cao, W., J. Mays, J. Dunn, R. Fulton, R. Silva, and A.
reticuloendotheliosis and Rous sarcoma. Avian Dis. Fadly. 2013. Use of polymerase chain reaction in
17:145–150. detection of Marek’s disease and reticuloendotheliosis
25 Beemon, K.L, A.J Faras, A.T Hasse, P.H Duesberg, and viruses in formalin‐fixed, paraffin‐embedded tumorous
J.E Maisel. 1976. Genomic complexities of murine tissues. Avian Dis. 57:785–789.
leukemia and sarcoma, reticuloendotheliosis, and visna 39 Carlson, H.C, G.L Seawright, and J.R Pettit. 1974.
viruses. J Virol. 17:525–537. Reticuloendotheliosis in Japanese quail. Avian Pathol.
26 Blank, V., P. Kourilsky, and A. Israel. 1992. NF‐kB and 3:169–175.
related proteins: Rel/dorsal homologies meet ankyrin‐ 40 Carpenter, C.R, H.R Bose, and A.S Rubin. 1977.
like repeats. Trends Biochem Sci. 17:135–140. Contact‐mediated suppression of mitogen‐induced
27 Bohls, R.L, E.W Collisson, S.L Gross, N.J Silvy, and D.N responsiveness by spleen cells in reticuloendotheliosis
Phalen. 2006. Experimental infection of Attwater’s/ virus‐induced tumorigenesis. Cell Immunol. 33:392–401.
greater prairie chicken hybrids with the 41 Carpenter, C.R, K.E Kempf, H.R Bose, and A.S Rubin.
reticuloendotheliosis virus. Avian Dis. 50:613–619. 1978. Characterization of the interaction of
28 Bohls, R.L, J.A Linares, S.L Gross, P.J Ferro, N.J Silvy, reticuloendotheliosis virus with the avian lymphoid
and E.W Collisson. 2006. Phylogenetic analyses indicate system. Cell Immunol. 39:307–315.
little variation among reticuloendotheliosis viruses 42 Carpenter, C.R, A.S Rubin, and H.R Bose, Jr. 1978.
infecting avian species, including the endangered Suppression of the mitogen‐stimulated blastogenic
Attwater’s prairie chicken. Virus Res. 119:187–194. response during reticuloendotheliosis virus‐induced
29 Bose, H.R, Jr. The 1992. Rel family: models for tumorigenesis: investigations into the mechanism of
transcriptional regulation and oncogenic action of the suppressor. J Immunol. 120:1313–1320.
transformation. Biochim Biophys Acta. 1114:1–17. 43 Casella, C.R, and A.T Panganiban. 1993. The matrix
30 Bose, H.R, Jr., and A.S Levine. 1967. Replication of the region is responsible for the differential ability of two
reticuloendotheliosis virus (strain T) in chicken retroviruses to function as helpers for vector
embryo cell culture. J Virol. 1:1117–1121. propagation. Virology. 192:458–464.
31 Breitman, M.L, M.M Lai, and P.K Vogt. 1980. 44 Chen, I.S, T.W Mak, J.J O’Rear, and H.M Temin. 1981.
Attenuation of avian reticuloendotheliosis virus: loss of Characterization of reticuloendotheliosis virus strain T
the defective transforming component during serial DNA and isolation of a novel variant of
passage of oncogenic virus in fibroblasts. Virology. reticuloendotheliosis virus strain T by molecular
101:304–306. cloning. J Virol. 40:800–811.
32 Bulow, V.V 1977. Immunological effects of 45 Chen, I.S, K.C Wilhelmsen, and H.M Temin. 1983.
reticuloendotheliosis virus as potential contaminant of Structure and expression of c‐rel, the cellular homolog
Marek’s disease vaccines. Avian Pathol. 6:383–393. to the oncogene of reticuloendotheliosis virus strain T.
33 Bulow, V.V 1980. Effects of infectious bursal disease J Virol. 45:104–113.
virus and reticuloendotheliosis virus infection of 46 Chen, P.Y, Z. Cui, L.F Lee, and R.L Witter. 1987.
chickens on the incidence of Marek’s disease and on Serologic differences among nondefective
local tumour development of the non‐producer JMV reticuloendotheliosis viruses. Arch Virol. 93:233–245.
transplant. Avian Pathol. 9:109–119. 47 Cho, B.R 1983. Cytopathic effects and focus formation
34 Bulow, V.V, and F. Weiland. 1980. Stimulation of local by reticuloendotheliosis viruses in a quail fibroblast cell
solid tumour development of the nonproducer Marek’s line. Avian Dis. 27:261–270.
disease tumour transplant JMV by virus‐induced 48 Cho, B.R 1984. Improved focus assay of
immunosuppression. Avian Pathol. 9:93–108. reticuloendotheliosis virus in a quail fibroblast cell line
35 Calvert, J.G, and K. Nazerian. 1994. An (QT35). Avian Dis. 28:261–265.
immunoperoxidase plaque assay for 49 Choi, K.M, and E.S Johnson. 2011. Industrial hygiene
reticuloendotheliosis virus and its application to a assessment of reticuloendotheliosis viruses exposure in
sensitive serum neutralization assay. Avian Dis. the poultry industry. Int Arch Occup Environ Health.
38:165–171. 84:375–382.
36 Calvert, J.G, K. Nazerian, R.L Witter, and N. Yanagida. 50 Coffin, J.M, R. Weiss, N. Teich, H. Varmus, and J.M
1993. Fowlpox virus recombinants expressing the Coffin. 1982. Structure of the retroviral genome. RNA
Chapter 15  Neoplastic Diseases 703

tumor viruses. In: Molecular Biology of Tumor Viruses. products of the reticuloendotheliosis proviral env and
J.M Coffin, ed. Cold Spring Harbor Laboratory, Cold LTR genes using psoralen‐biotin labelled probes. J Virol
Spring Harbor, NY. 261–368. Methods. 59:113–119.
51 Cohen, R.S, T.C Wong, and M.M Lai. 1981. 65 Davidson, I., S. Perl, and M. Malkinson. 1998. A 4‐year
Characterization of transformation‐ and replication‐ survey of avian oncogenic viruses in tumour‐bearing
specific sequences of reticuloendotheliosis virus. flocks in Israel—a comparison of PCR, serology and
Virology. 113:672–685. histopathology. Avian Pathol. 27:S90–S90.
52 Cook, M.K 1969. Cultivation of filterable agent 66 Deng, X., X. Qi, Y. Gao, Y. Wang, L. Qin, H. Gao, L.
associated with Marek’s disease. J Nat Cancer Instit. Gao, and X. Wang. 2010. Development of a loop‐
43:203–212. mediated isothermal amplification method for rapid
53 Cooper, M.D, C.L Chen, R.P Bucy, and C.B Thompson. detection of reticuloendotheliosis virus. J Virol
Avian 1991. T cell ontogeny. Adv Immunol. 50:87–117. Methods. 168:82–86.
54 Cowen, B.S, and M.O Braune. 1988. The propagation of 67 Diallo, I.S, M.A Mackenzie, P.B Spradbrow, and W.F
avian viruses in a continuous cell line (QT35) of Robinson. 1998. Field isolates of fowlpox virus
Japanese quail origin. Avian Dis. 32:282–297. contaminated with reticuloendotheliosis virus. Avian
55 Crespo, R., P.R Woolcock, A.M Fadly, C. Hall, and H.L Pathol. 27:60–66.
Shivaprasad. 2002. Characterization of T‐cell 68 DiGiacomo, R.F, and S.G Hopkins. 1997. Food animal
lymphomas associated with an outbreak of and poultry retroviruses and human health. Vet Clin
reticuloendotheliosis in turkeys. Avian Pathol. 31:355–361. North Am Food Anim Pract. 13:177–190.
56 Crittenden, L.B, A.M Fadly, and E.J Smith. 1982. Effect 69 Dimier, I.H, P. Quere, M. Naciri, and D.T Bout. 1998.
of endogenous leukosis virus genes on response to Inhibition of Eimeria tenella development in vitro
infection with avian leukosis and reticuloendotheliosis mediated by chicken macrophages and fibroblasts
viruses. Avian Dis. 26:279–294. treated with chicken cell supernatants with IFN‐gamma
57 Cui, Z.Z, L.F Lee, R.F Silva, and R.L Witter. 1986. activity. Avian Dis. 42:239–247.
Monoclonal antibodies against avian reticuloendotheliosis 70 Dornburg, R. 1995. Reticuloendotheliosis viruses and
virus: identification of strain‐specific and strain‐common derived vectors. Gene Ther. 2:301–310.
epitopes. J Immunol. 136:4237–4242. 71 Dornburg, R. 2003. Reticuloendotheliosis viruses and
58 Cui, Z.Z, L.F Lee, E.J Smith, R.L Witter, and T.S Chang. derived vectors for human gene therapy. Front Biosci.
1988. Monoclonal‐antibody‐mediated enzyme‐linked 8:d801–817.
immunosorbent assay for detection of 72 Dren, C.N, I. Nemeth, I. Sari, F. Ratz, R. Glavits, and
reticuloendotheliosis viruses. Avian Dis. 32:32–40. P. Somogyi. 1988. Isolation of a reticuloendotheliosis‐
59 Davidson, I., R. Alphandary, M. Novoseler, and M. like virus from naturally occurring lymphoreticular
Malkinson. 1997. Replication of non‐defective tumours of domestic goose. Avian Pathol.
reticuloendotheliosis viruses in the avian embryo 17:259–277.
assayed by PCR and immunofluorescence. Avian 73 Dren, C.N, E. Saghy, R. Glavits, F. Ratz, J. Ping, and V.
Pathol. 26:579–593. Sztojkov. 1983. Lymphoreticular tumour in pen‐raised
60 Davidson, I., and R. Borenshtain. 2001. In vivo events of pheasants associated with a reticuloendotheliosis like
retroviral long terminal repeat integration into Marek’s virus infection. Avian Pathol. 12:55–71.
disease virus in commercial poultry: detection of 74 Drew, M.L, W.L Wigle, D.L Graham, C.P Griffin, N.J
chimeric molecules as a marker. Avian Dis. 45:102–121. Silvy, A.M Fadly, and R.L Witter. 1998.
61 Davidson, I., A. Borovskaya, S. Perl, and M. Malkinson. Reticuloendotheliosis in captive greater and Attwater’s
1995. Use of the polymerase chain reaction for the prairie chickens. J Wildl Dis. 34:783–791.
diagnosis of natural infection of chickens and turkeys 75 Embretson, J.E, and H.M Temin. 1986. Pseudotyped
with Marek’s disease virus and reticuloendotheliosis retroviral vectors reveal restrictions to
virus. Avian Pathol. 24:69–94. reticuloendotheliosis virus replication in rat cells.
62 Davidson, I., and Y. Braverman. 2005. Insect contribution J Virol. 60:662–668.
to horizontal transmission of Reticuloendotheliosis 76 Embretson, J.E, and H.M Temin. 1987. Transcription
virus. J Med Entomol. 42:128–133. from a spleen necrosis virus 5′ long terminal repeat is
63 Davidson, I., A.M Fadly, K.A Schat, and J.L Spencer. suppressed in mouse cells. J Virol. 61:3454–3462.
1997. Epidemiology and control of REV in chickens and 77 Etienne, L., and M. Emerman. 2013. The mongoose, the
turkeys in Israel. In: Diagnosis and Control of pheasant, the pox, and the retrovirus. PLoS Biol.
Neoplastic Diseases. American Association of Avian 11:e1001641.
Pathologists, Kennet Square, PA. 70–75. 78 Fadly, A., and M.C Garcia. 2006. Detection of
64 Davidson, I., and M. Malkinson. 1996. A non‐ reticuloendotheliosis virus in live virus vaccines of
radioactive method for identifying enzyme‐amplified poultry. Dev Biol (Basel). 126:301–305; discussion 327.
704 Section II  Viral Diseases

79 Fadly, A.M 1993. Induction of antibodies to avian 92 Garcia, M., N. Narang, W.M Reed, and A.M Fadly.
leukosis and reticuloendotheliosis viruses using 2003. Molecular characterization of
defective retroviral particles. In: 130th American reticuloendotheliosis virus insertions in the genome
Veterinary Medical Association. Minneapolis, MN. of field and vaccine strains of fowl poxvirus. Avian
80 Fadly, A.M, M.L Drew, and R.L Witter. 1996. Isolation Dis. 47:343–354.
of a Nondefective Strain of Reticuloendotheliosis Virus 93 Garry, R.F, G.M Shackleford, L.J Berry, and H.R Bose,
from Greater and Attwater’s Prairie Chickens. In: 45th Jr. 1985. Inhibition of hepatic phosphoenolpyruvate
Western Poultry Disease Conference. Sacramento, CA. carboxykinase by avian reticuloendotheliosis viruses.
317–318. Cancer Res. 45:5020–5026.
81 Fadly, A.M, K.A Schat, and J.L Spencer. 1997. Criteria 94 Gautier, R., A. Jiang, V. Rousseau, R. Dornburg, and T.
for the differential diagnosis of viral lymphomas of Jaffredo. 2000. Avian reticuloendotheliosis virus strain
chickens: a review. In: Diagnosis and Control of A and spleen necrosis virus do not infect human cells.
Neoplastic Diseases of Poultry. American Association J Virol. 74:518–522.
of Avian Pathologists, Kennet Square. 6–11. 95 Genovese, K.J, R.B Moyes, L.L Genovese, V.K Lowry,
82 Fadly, A.M, and R.L Witter. 1983. Studies of and M.H Kogut. 1998. Resistance to Salmonella
reticuloendotheliosis virus‐induced lymphomagenesis enteritidis organ invasion in day‐old turkeys and
in chickens. Avian Dis. 27:271–282. chickens by transformed T‐cell line‐produced
83 Fadly, A.M, and R.L Witter. 1986. Resistance of line 6(3) lymphokines. Avian Dis. 42:545–553.
chickens to reticuloendotheliosis‐virus‐induced 96 Grimes, T.M, T.J Bagust, and C.K Dimmock. 1979.
bursa‐associated lymphomas. Int J Cancer. 38:139–143. Experimental infection of chickens with an australian
84 Fadly, A.M, and R.L Witter. 1997. Comparative strain of reticuloendotheliosis virus. I. Clinical,
evaluation of in vitro and in vivo assays for the pathological and haematological effects. Avian Pathol.
detection of reticuloendotheliosis virus as a 8:57–68.
contaminant in a live virus vaccine of poultry. Avian 97 Grimes, T.M, and H.G Purchase. 1973.
Dis. 41:695–701. Reticuloendotheliosis in a duck. Aust Vet J. 49:466–471.
85 Fadly, A.M, R.L Witter, E.J Smith, R.F Silva, W.M 98 Hafner, S., M.A Goodwin, E.J Smith, D.I Bounous, M.
Reed, F.J Hoerr, and M.R Putnam. 1996. An outbreak Puette, L.C Kelley, K.A Langheinrich, and A.M Fadly.
of lymphomas in commercial broiler breeder 1996. Multicentric histiocytosis in young chickens.
chickens vaccinated with a fowlpox vaccine Gross and light microscopic pathology. Avian Dis.
contaminated with reticuloendotheliosis virus. Avian 40:202–209.
Pathol. 25:35–47. 99 Hayes, L.E, K.A Langheinrich, and R.L Witter. 1992.
86 Federspiel, M.J, L.B Crittenden, and S.H Hughes. 1989. Reticuloendotheliosis in a wild turkey (Meleagris
Expression of avian reticuloendotheliosis virus gallopavo) from coastal Georgia. J Wildl Dis.
envelope confers host resistance. Virology. 173:167–177. 28:154–158.
87 Ferro, P.J, M.E Morrow, J.P Flanagan, B. Ortego, R.E 100 Hertig, C., B.E Coupar, A.R Gould, and D.B Boyle.
Chester, J.M Mueller, and B. Lupiani. Wild 2017. Birds, 1997. Field and vaccine strains of fowlpox virus carry
a source of reticuloendotheliosis virus infection for the integrated sequences from the avian retrovirus,
endangered Attwater’s prairie‐chicken (Tympanuchus reticuloendotheliosis virus. Virology. 235:367–376.
cupido attwateri)? J Wildl Dis. 53:586–590. 101 Hoelzer, J.D, R.B Franklin, and H.R Bose, Jr. 1979.
88 Filardo, E.J, M.F Lee, and E.H Humphries. 1994. Transformation by reticuloendotheliosis virus:
Structural genes, not the LTRs, are the primary development of a focus assay and isolation of a
determinants of reticuloendotheliosis virus A‐induced nontransforming virus. Virology. 93:20–30.
runting and bursal atrophy. Virology. 202:116–128. 102 Hoelzer, J.D, R.B Lewis, C.R Wasmuth, and H.R Bose,
89 Franklin, R.B, C.Y Kang, K. Min‐Min Wan, and H.R Jr. 1980. Hematopoietic cell transformation by
Bose, Jr. 1977. Transformation of chick embryo reticuloendotheliosis virus: characterization of the
fibroblasts by reticuloendotheliosis virus. Virology. genetic defect. Virology. 100:462–474.
83:313–321. 103 Hrdlickova, R., J. Nehyba, and E.H Humphries. 1994.
90 Fritsch, E., and H.M Temin. 1977. Formation and v‐rel induces expression of three avian
structure of infectious DNA of spleen necrosis virus. immunoregulatory surface receptors more efficiently
J Virol. 21:119–130. than c‐rel. J Virol. 68:308–319.
91 Fujita, D.J, R.A Swift, A.G Ridgway, and H.J Kung. 104 Hu, C., and T.J Linna. 1976. Serotherapy of avian
1984. Reticuloendotheliosis virus induced B reticuloendotheliosis virus‐induced tumors. Ann N Y
lymphomas in chickens characterization of a tumour Acad Sci. 277:634–646.
cell DNA clone containing proviral and c‐myc 105 Humphries, E.H, and G. Zhang. 1992. V‐rel and
sequences. J Cell Biochem. 7(B):12–12. C‐rel modulate the expression of both bursal and
Chapter 15  Neoplastic Diseases 705

non‐bursal antigens on avian B‐cell lymphomas. Curr 119 Kang, C.Y, T.C Wong, and K.V Holmes. 1975.
Top Microbiol Immunol. 182:475–483. Comparative ultrastructural study of four
106 Ianconescu, M. 1977. Reticuloendotheliosis antigen reticuloendothelias viruses. J Virol. 16:1027–1038.
for the agar gel precipitation test. Avian Pathol. 120 Kawamura, H., T. Wakabayashi, S. Yamaguchi, T.
6:259–267. Taniguchi, and N. Takayanagi. 1976. Inoculation
107 Ianconescu, M., and A. Aharonovici. 1978. Persistent experiment of Marek’s disease vaccine contaminated
viraemia in chickens subsequent to in ovo inoculation with a reticuloendotheliosis virus. Natl Inst Anim
of reticuloendotheliosis virus. Avian Pathol. Health Q (Tokyo). 16:135–140.
7:237–247. 121 Keshet, E., and H.M Temin. 1979. Cell. killing by
108 Ignjatovic, J., K.J Fahey, and T.J Bagust. 1987. An spleen necrosis virus is correlated with a transient
enzyme‐linked immunosorbent assay for detection of accumulation of spleen necrosis virus DNA. J Virol.
reticuloendotheliosis virus infection in chickens. 31:376–388.
Avian Pathol. 16:609–621. 122 Kewalramani, V.N, A.T Panganiban, and M.
109 Isfort, R., R.L Witter, and H.J Kung. 1987. C‐myc Emerman. 1992. Spleen necrosis virus, an avian
activation in an unusual retrovirus‐induced avian immunosuppressive retrovirus, shares a receptor with
T‐lymphoma resembling Marek’s disease: proviral the type D simian retroviruses. J Virol. 66:3026–3031.
insertion 5′ of exon one enhances the expression of an 123 Kim, T.J, and D.N Tripathy. 2001.
intron promoter. Oncogene. Res 2:81–94. Reticuloendotheliosis virus integration in the fowl
110 Isfort, R.J, Z. Qian, D. Jones, R.F Silva, R. Witter, and poxvirus genome: not a recent event. Avian Dis.
H.J Kung. 1994. Integration of multiple chicken 45:663–669.
retroviruses into multiple chicken herpesviruses: 124 Kochel, T., and N.R Rice. 1992. v‐rel‐ and c‐rel‐protein
herpesviral gD as a common target of integration. complexes bind to the NF‐kappa B site in vitro.
Virology. 203:125–133. Oncogene. 7:567–572.
111 Isfort, R.J, R. Witter, and H.J Kung. 1994. Retrovirus 125 Koo, H.M, A.M Brown, Y. Ron, and J.P Dougherty.
insertion into herpesviruses. Trends Microbiol. 1991. Spleen necrosis virus, an avian retrovirus, can
2:174–177. infect primate cells. J Virol. 65:4769–4776.
112 Jackson, C.A, S.E Dunn, D.I Smith, P.T Gilchrist, and 126 Koo, H.M, J. Gu, A. Varela‐Echavarria, Y. Ron, and J.P
P.A Macqueen. 1977. Proventriculitis, “nakanuke” and Dougherty. 1992. Reticuloendotheliosis type C and
reticuloendotheliosis in chickens following primate type D oncoretroviruses are members of the
vaccination with herpesvirus of turkeys (HVT). Aust same receptor interference group. J Virol. 66:3448–3454.
Vet J. 53:457–459. 127 Koyama, H., K. Inoue, T. Nagashima, Y. Ohwada, and
113 Johnson, E.S 1994. Poultry oncogenic retroviruses and Y. Saito. 1975. Cause of “nakanuke” in chickens. I.
humans. Cancer Detect Prev. 18:9–30. Occurrence of “nakanuke” in chicken inoculated with
114 Johnson, E.S, L.G Nicholson, and D.T Durack. 1995. the cells showed coexistence or C‐type virus and
Detection of antibodies to avian leukosis/sarcoma turkey herpesvirus. Kitasato Arch Exp Med. 48:83–91.
viruses (ALSV) and reticuloendotheliosis viruses 128 Koyama, H., T. Nagashima, Y. Ohwada, and Y. Saito.
(REV) in humans by ELISA. Cancer Detect Prev. 1975. Cause of “nakanuke” in chickens. II. Isolation of
19:394–404. C‐type virus from material infected with turkey
115 Johnson, E.S, L. Overby, and R. Philpot. 1995. herpesvirus. Kitasato Arch Exp Med. 48:93–105.
Detection of antibodies to avian leukosis/sarcoma 129 Koyama, H., T. Sasaki, Y. Ohwada, and Y. Saito. 1980.
viruses and reticuloendotheliosis viruses in humans The relationship between feathering abnormalities
by western blot assay. Cancer Detect Prev. (“nakanuke”) and tumour production in chickens
19:472–486. inoculated with reticuloendotheliosis virus. Avian
116 Kang, C.Y 1975. Characterization of endogenous Pathol. 9:331–340.
RNA‐directed DNA polymerase activity of 130 Larose, R.N, and M. Sevoian. 1965. Avian
reticuloendotheliosis viruses. J Virol. 16:880–886. lymphomatosis. IX. Mortality and serological
117 Kang, C.Y, and P. Lambright. 1977. Pseudotypes of response of chickens of various ages to graded doses
vesicular stomatitis virus with the mixed coat of of T strain. Avian Dis. 9:604–610.
reticuloendotheliosis virus and vesicular stomatitis 131 Lewis, R.B, J. McClure, B. Rup, D.W Niesel, R.F Garry,
virus. J Virol. 21:1252–1255. J.D Hoelzer, K. Nazerian, and H.R Bose, Jr. 1981.
118 Kang, C.Y, and H.M Temin. 1973. Lack of sequence Avian reticuloendotheliosis virus: identification of the
homology among RNAs of avian leukosis‐sarcoma hematopoietic target cell for transformation. Cell.
viruses, reticuloendotheliosis viruses, and chicken 25:421–431.
endogenous RNA‐directed DNA polymerase activity. 132 Ley, D.H, M.D Ficken, D.T Cobb, and R.L Witter.
J Virol. 12:1314–1324. 1989. Histomoniasis and reticuloendotheliosis in a
706 Section II  Viral Diseases

wild turkey (Meleagris gallopavo) in North Carolina. 147 McDougall, J.S, J.B McFerran, and M.S McNulty.
J  Wildl Dis. 25:262–265. 1993. Tumor viruses of turkeys. In: Virus Infections of
133 Li, J., B.W Calnek, K.A Schat, and D.L Graham. 1983. Vertebrates. Elsevier Science Publishers, Amsterdam.
Pathogenesis of reticuloendotheliosis virus infection 455–463.
in ducks. Avian Dis. 27:1090–1105. 148 McDougall, J.S, R.W Shilleto, and P.M Biggs. 1980.
134 Li, J., X. Dong, C. Yang, Q. Li, Z. Cui, S. Chang, P. Experimental infection and vertical transmission of
Zhao, K. Yu, and H. Yang. 2015. Isolation, reticuloendotheliosis virus in the turkey. Avian Pathol.
identification, and whole genome sequencing of 9:445–454.
reticuloendotheliosis virus from a vaccine against 149 McDougall, J.S, R.W Shilleto, and P.M Biggs. 1981.
Marek’s disease. Poult Sci. 94:643–649. Further studies on vertical transmission of
135 Li, J., C. Yang, Q. Li, H. Li, Y. Xia, D. Liu, K. Yu, and H. reticuloendotheliosis virus in turkeys. Avian Pathol.
Yang. 2013. Complete genome sequence of 10:163–169.
reticuloendotheliosis virus strain MD‐2, isolated from 150 Meroz, M. 1992. Reticuloendotheliosis and “pullet
a contaminated turkey herpesvirus vaccine. Genome disease” in Israel. Vet Rec. 130:107–108.
Announc. 1. 151 Meyers, N.L 1993. Antibody response elicited against
136 Lim, M.Y, N. Davis, J.Y Zhang, and H.R Bose, Jr. 1990. empty reticuloendotheliosis virus particles in two
The v‐rel oncogene product is complexed with cellular inbred lines of chicken. Vet Microbiol. 36:317–332.
proteins including its proto‐oncogene product and 152 Miller, P.E, J. Paul‐Murphy, R. Sullivan, A.J Cooley,
heat shock protein 70. Virology. 175:149–160. R.R Dubielzig, C.J Murphy, and A.M Fadly. 1998.
137 Linna, T.J, C.P Hu, and K.D Thompson. 1974. Orbital lymphosarcoma associated with
Development of systemic and local tumors induced by reticuloendotheliosis virus in a peafowl. J Am Vet Med
avian reticuloendotheliosis virus after thymectomy or Assoc. 213:377–380.
bursectomy. J Natl Cancer Inst. 53:847–854. 153 Moelling, K., H. Gelderblom, G. Pauli, and R. Friis.
138 Liu, Q., J. Zhao, J. Su, J. Pu, G. Zhang, and J. Liu. 2009. 1975. A comparative study of the avian
Full genome sequences of two reticuloendotheliosis reticuloendotheliosis virus: relationship to murine
viruses contaminating commercial vaccines. Avian leukemia virus and viruses of the avian sarcoma‐
Dis. 53:341–346. leukosis complex. Virology. 65:546–557.
139 Luan, H., Y. Wang, Y. Li, Z. Cui, S. Chang, and P. Zhao. 154 Moore, K.M, J.R Davis, T. Sato, and A. Yasuda. 2000.
2016. Development of a real‐time quantitative RT‐ Reticuloendotheliosis virus (REV) long terminal
PCR to detect REV contamination in live vaccine. repeats incorporated in the genomes of commercial
Poult Sci. 95:2023–2029. fowl poxvirus vaccines and pigeon poxviruses without
140 Ludford, C.G, H.G Purchase, and H.W Cox. 1972. indication of the presence of infectious REV. Avian
Duck infectious anemia virus associated with Dis. 44:827–841.
Plasmodium lophurae. Exp Parasitol. 31:29–38. 155 Moscovici, C., D. Chi, L. Gazzolo, and M.G
141 Maccubbin, D.L, and L.W Schierman. 1986. MHC‐ Moscovici. 1976. A study of plaque formation with
restricted cytotoxic response of chicken T cells: avian RNA tumor viruses. Virology. 73:181–189.
expression, augmentation, and clonal characterization. 156 Mosser, A.G 1975. Polypeptide composition of spleen
J Immunol. 136:12–16. necrosis virus, a reticuloendotheliosis virus. J Virol.
142 Maldonado, R.L, and H.R Bose. 1971. Separation of 15:1088–1095.
reticuloendotheliosis virus from avian tumor viruses. 157 Motha, M.X 1982. Effects of reticuloendotheliosis
J Virol. 8:813–815. virus on the response of chickens to infectious
143 Maldonado, R.L, and H.R Bose, Jr. 1976. Group‐ laryngotracheitis virus. Avian Pathol. 11:475–486.
specific antigen shared by the members of the 158 Motha, M.X 1984. Distribution of virus and tumour
reticuloendotheliosis virus complex. J Virol. 17:983–990. formation in ducks experimentally infected with
144 Martinez, I., and R. Dornburg. 1996. Mutational reticuloendotheliosis virus. Avian Pathol. 13:303–319.
analysis of the envelope protein of spleen necrosis 159 Motha, M.X 1987. Clinical effects, virological and
virus. J Virol. 70:6036–6043. serological responses in chickens following in‐ovo
145 McColl, K.A, B.W Calnek, W.V Harris, K.A Schat, and inoculation of reticuloendotheliosis virus. Vet
L.F Lee. 1987. Expression of a putative tumor‐ Microbiol. 14:411–417.
associated surface antigen on normal versus Marek’s 160 Motha, M.X, and J.R Egerton. 1983. Effect of
disease virus‐transformed lymphocytes. J Natl Cancer reticuloendotheliosis virus on the response of
Inst. 79:991–1000. chickens to Salmonella typhimurium infection. Res
146 McDougall, J.S, P.M Biggs, and R.W Shilleto. 1978. A Vet Sci. 34:188–192.
leukosis in turkeys associated with infection with 161 Motha, M.X, and J.R Egerton. 1984. Influence of
reticuloendotheliosis virus. Avian Pathol. 7:557–568. reticuloendotheliosis on the severity of Eimeria tenella
Chapter 15  Neoplastic Diseases 707

infection in broiler chickens. Vet Microbiol. 175 Paul, P.S, K.A Pomeroy, P.S Sarma, K.H Johnson, D.M
9:121–129. Barnes, M.C Kumar, and B.S Pomeroy. 1976. Naturally
162 Motha, M.X, and J.R Egerton. 1987. Outbreak of occurring reticuloendotheliosis in turkeys:
atypical fowlpox in chickens with persistent transmission. J Natl Cancer Inst. 56:419–422.
reticuloendotheliosis viraemia. Avian Pathol. 176 Paul, P.S, R.E Werdin, and B.S Pomeroy. 1978.
16:177–182. Spontaneously occurring lymphoproliferative disease
163 Motha, M.X, and J.R Egerton. 1987. Vertical in ducks. Avian Dis. 22:191–195.
transmission of reticuloendotheliosis virus in 177 Payne, L.N 1992. Biology of avian retroviruses. In:
chickens. Avian Pathol. 16:141–147. Retroviridae. Plenum Press, New York. 299–404.
164 Motha, M.X, J.R Egerton, and A.W Sweeney. 1984. 178 Perk, K., A. Malkinson, A. Gazit, A. Yaniv, and A.
Some evidence of mechanical transmission of Zimbber. 1981. Reappearance of an acute
reticuloendotheliosis virus by mosquitoes. Avian Dis. undifferentiated leukemia in a flock of Muscovy
28:858–867. ducks. In: 10th International Symposium on
165 Mussman, H.C, and M.J Twiehaus. 1971. Pathogenesis Comparative Leukemia and Related Diseases. 99–100.
of reticuloendothelial virus disease in chicks–an acute 179 Peterson, D.A, and A.S Levine. 1971. Avian
runting syndrome. Avian Dis. 15:483–502. reticuloendotheliosis virus (strain T). IV. Infectivity
166 Nair, V., G. Zavala, and A.M Fadly. and transmissibility in day‐old cockerels. Avian Dis.
Reticuloendotheliosis. In: 2013. Diseases of Poultry, 15:874–883.
13th ed. D.E Swayne, J.R Glisson, L.R McDougald, L.K 180 Peterson, M.J, P.J Ferro, M.N Peterson, R.M Sullivan,
Nolan, D.L Suarez and V. Nair, eds. Wiley‐Blackwell, B.E Toole, and N.J Silvy. 2002. Infectious disease
Ames, IA. 593–604. survey of lesser prairie chickens in north Texas.
167 Nazerian, K., J.G Calvert, R.L Witter, and N. Yanagida. J Wildl Dis. 38:834–839.
1995. Vaccine comprising fowlpox virus recombinants 181 Pratt, W.D, R.W Morgan, and K.A Schat. 1992.
expressing the envelope glycoprotein of an avian Characterization of reticuloendotheliosis virus‐
reticuloendotheliosis retrovirus. In. U.S P. Office, ed., transformed avian T‐lymphoblastoid cell lines
U.S.A. infected with Marek’s disease virus. J Virol.
168 Nazerian, K., R.L Witter, L.B Crittenden, M.R 66:7239–7244.
Noori‐Dalloii, and H.J Kung. 1982. An IgM‐ 182 Purchase, H.G, C. Ludford, K. Nazerian, and H.W
producing B lymphoblastoid cell line established Cox. 1973. A new group of oncogenic viruses:
from lymphomas induced by a non‐defective reticuloendotheliosis, chick syncytial, duck infectious
reticuloendotheliosis virus. J Gen Virol. 58:351–360. anemia, and spleen necrosis viruses. J Natl Cancer
169 Nicholas, R.A, and D.H Thornton. 1983. Relative Inst. 51:489–499.
efficiency of techniques for detecting avian 183 Purchase, H.G, and R.L Witter. 1975. The
reticuloendotheliosis virus as a vaccine contaminant. reticuloendotheliosis viruses. Curr Top Microbiol
Res Vet Sci. 34:377–379. Immunol. 71:103–124.
170 Nicholas, R.A, and D.H Thornton. 1987. An enzyme‐ 184 Ratnamohan, N., T.J Bagust, and P.B Spradbrow. 1982.
linked immunosorbent assay for the detection of Establishment of a chicken lymphoblastoid cell line
antibodies to avian reticuloendotheliosis virus using infected with reticuloendotheliosis virus. J Comp
whole cell antigen. Res Vet Sci. 43:403–404. Pathol. 92:527–532.
171 Niewiadomska, A.M, and R.J Gifford. 2013. The 185 Ratnamohan, N., T.M Grimes, Bagust, and P.B
extraordinary evolutionary history of the Spradbrow. 1980. A transmissible chicken tumour
reticuloendotheliosis viruses. PLoS Biol. associated with reticuloendotheliosis virus infection.
11:e1001642. Aust Vet J. 56:34–38.
172 Noori‐Daloii, M.R, R.A Swift, H.J Kung, L.B 186 Ratnamohan, R., T.J Bagust, T.M Grimes, and P.B
Crittenden, and R.L Witter. 1981. Specific integration Spradbrow. 1979. Transmission of an Australian strain
of REV proviruses in avian bursal lymphomas. Nature. of reticuloendotheliosis virus to adult Japanese quail.
294:574–576. Aust Vet J. 55:506.
173 Okoye, J.O, W. Ezema, and J.N Agoha. 1993. Naturally 187 Reddy, S.K, M.J Ratcliffe, and A. Silim. 1993. Flow
occurring clinical reticuloendotheliosis in turkeys and cytometric analysis of the neutralizing immune
chickens. Avian Pathol. 22:237–244. response against infectious bursal disease virus using
174 Paul, P.S, K.H Johnson, K.A Pomeroy, B.S Pomeroy, reticuloendotheliosis virus‐transformed
and P.S Sarma. 1977. Experimental transmission of lymphoblastoid cell lines. J Virol. Methods
reticuloendotheliosis in turkeys with the cell‐culture‐ 44:167–177.
propagated reticuloendotheliosis viruses of turkey 188 Reddy, S.M, R.L Witter, and I. Gimeno. 2000.
origin. J Natl Cancer Inst. 58:1819–1824. Development of a quantitative‐competitive
708 Section II  Viral Diseases

polymerase chain reaction assay for serotype 1 202 Schat, K.A, W.D Pratt, R. Morgan, D. Weinstock, and
Marek’s disease virus. Avian Dis. 44:770–775. B.W Calnek. 1992. Stable transfection of
189 Rice, N.R, T.I Bonner, and R.V Gilden. 1981. Nucleic reticuloendotheliosis virus‐transformed
acid homology between avian and mammalian type C lymphoblastoid cell lines. Avian Dis. 36:432–439.
viruses: relatedness of reticuloendotheliosis virus cdna 203 Schwarzbard, Z., A. Yaniv, M. Ianconescu, K. Perk,
to cloned proviral DNA of the endogenous Colobus and A. Zimber. 1980. A reverse transcriptase assay for
virus CPC‐1. Virology. 114:286–290. the diagnosis of lymphoproliferative disease (LPD) of
190 Ridgway, A.A, R.A Swift, H.J Kung, and D.J Fujita. turkeys. Avian Pathol. 9:481–487.
1985. In vitro transcription analysis of the viral 204 Scofield, V.L, and H.R Bose, Jr. 1978. Depression of
promoter involved in c‐myc activation in chicken B mitogen response in spleen cells from
lymphomas: detection and mapping of two RNA reticuloendotheliosis virus‐infected chickens and their
initiation sites within the reticuloendotheliosis virus suppressive effect on normal lymphocyte response.
long terminal repeat. J Virol. 54:161–170. J Immunol. 120:1321–1325.
191 Robinson, F.R, and M.J Twiehaus. 1974. Isolation of 205 Scofield, V.L, J.L Spence, W.E Briles, and H.R Bose, Jr.
the avian reticuloendotheliosis virus (strain T). Avian 1978. Differential mortality and lesion responses to
Dis. 18:278–288. reticuloendotheliosis virus infection in Marek’s
192 Rup, B.J, J.D Hoelzer, and H.R Bose, Jr. 1982. Helper disease‐resistant and susceptible chicken lines.
viruses associated with avian acute leukemia viruses Immunogenetics. 7:169–172.
inhibit the cellular immune response. Virology. 116:61–71. 206 Seong, H.W, S.J Kim, J.H Kim, C.S Song, I.P Mo, and
193 Rup, B.J, J.L Spence, J.D Hoelzer, R.B Lewis, C.R K.S Kim. 1996. Outbreaks of reticuloendotheliosis in
Carpenter, A.S Rubin, and H.R Bose, Jr. 1979. Korea. RDA J Agric Sci. 38:707–715.
Immunosuppression induced by avian 207 Sevoian, M. 1964. Avian lymphomatosis. VI. A virus
reticuloendotheliosis virus: mechanism of induction of unusual potency and pathogenicity. Avian Dis.
of the suppressor cell. J Immunol. 123:1362–1370. 3:336–347.
194 Rushlow, C., and R. Warrior. 1992. The rel family of 208 Sevoian, M., R.N Larose, and D.M Chamberlain. 1964.
proteins. Bioessays. 14:89–95. Pathological response of the chicken embryo to T
195 Salter, D.W, E.J Smith, S.H Hughes, S.E Wright, and virus. J Nat Cancer Instit. 17:99–119.
L.B Crittenden. 1987. Transgenic chickens: insertion 209 Simek, S., and N.R Rice. 1980. Analysis of the nucleic
of retroviral genes into the chicken germ line. acid components in reticuloendotheliosis virus. J
Virology. 157:236–240. Virol. 33:320–329.
196 Santos, V.L, S.M Williams, G. Zavala, T. Barbosa, J. 210 Singh, P., T.J Kim, and D.N Tripathy. 2000. Re‐
Zhang, S. Cheng, H.L Shivaprasad, S. Hafner, A. Fadly, emerging fowlpox: evaluation of isolates from
R.L Santos, and C.C Brown. 2008. Detection of vaccinated flocks. Avian Pathol. 29:449–455.
reticuloendotheliosis virus by immunohistochemistry 211 Singh, P., W.M Schnitzlein, and D.N Tripathy. 2003.
and in situ hybridization in experimentally infected Reticuloendotheliosis virus sequences within the
Japanese quail embryos and archived formalin‐fixed genomes of field strains of fowlpox virus display
and paraffin‐embedded tumours. Avian Pathol. variability. J Virol. 77:5855–5862.
37:451–456. 212 Sinkovic, B. 1981. In vivo interactions between
197 Sarid, R., A. Chajut, M. Malkinson, S.R Tronick, A. reticuloendotheliosis virus and some other infectious
Gazit, and A. Yaniv. 1994. Diagnostic test for agents of chickens. In: 4th Australasian Poultry Stock
lymphoproliferative disease virus infection of turkeys, and Feed Convention. 114–118.
using the polymerase chain reaction. Am J Vet Res. 213 Sinkovic, B., and O. Choi. 1979. Studies on
55:769–772. reticuloendotheliosis maternal antibody. In: 3rd
198 Sawyer, R.C, and H. Hanafusa. 1977. Formation of Australasian Poultry Stock and Feed Convention.
reticuloendotheliosis virus pseudotypes of Rous 119–122.
sarcoma virus. J Virol. 22:634–639. 214 Smith, E.J, J.J Solomon, and R.L Witter. 1977.
199 Schat, K.A 1991. Importance of cell‐mediated Complement‐fixation test for reticuloendotheliosis
immunity in Marek’s disease and other viral tumor viruses: limits of sensitivity in infected avian cells.
diseases. Poult Sci. 70:1165–1175. Avian Dis. 21:612–622.
200 Schat, K.A, and H.N Erb. 2014. Lack of evidence that 215 Solomon, J.J, R.L Witter, and K. Nazerian. 1976.
avian oncogenic viruses are infectious for humans: a Studies on the etiology of lymphomas in turkeys:
review. Avian Dis. 58:345–358. isolation of reticuloendotheliosis in virus. Avian Dis.
201 Schat, K.A, J. Gonzalez, A. Solorzano, E. Avila, and 20:735–747.
R.L Witter. 1976. A lymphoproliferative disease in 216 Storms, R.W, and H.R Bose, Jr. 1992. Alterations
Japanese Quail. Avian Dis. 20:153–161. within pp59v‐rel‐containing protein complexes
Chapter 15  Neoplastic Diseases 709

following the stimulation of REV‐T‐transformed 229 Thompson, K.D, R.G Fischer, and D.H Luecke. 1971.
lymphoid cells with zinc. Virology. 188:765–777. Quantitative infectivity studies of avian
217 Swift, R.A, C. Boerkoel, A. Ridgway, D.J Fujita, J.B reticuloendotheliosis virus (strain T) in certain
Dodgson, and H.J Kung. 1987. B‐lymphoma induction hematophagous arthropods. J Med Entomol. 8:486–490.
by reticuloendotheliosis virus: characterization of a 230 Trager, W. 1959. A new virus of ducks interfering with
mutated chicken syncytial virus provirus involved in development of malaria parasite (Plasmodium
c‐myc activation. J Virol. 61:2084–2090. lophurae). Proc Soc Exp Biol Med. 101:578–582.
218 Swift, R.A, E. Shaller, R.L Witter, and H.J Kung. 1985. 231 Trampel, D.W, T.M Pepper, and R.L Witter. 2002.
Insertional activation of c‐myc by Reticuloendotheliosis in Hungarian partridge. J Wildl
reticuloendotheliosis virus in chicken B lymphoma: Dis. 38:438–442.
nonrandom distribution and orientation of the 232 Tsai, W.P, T.D Copeland, and S. Oroszlan. 1985.
proviruses. J Virol. 54:869–872. Purification and chemical and immunological
219 Tajima, M., T. Nunoya, and Y. Otaki. 1977. characterization of avian reticuloendotheliosis virus
Pathogenesis of abnormal feathers in chickens gag‐gene‐encoded structural proteins. Virology.
inoculated with reticuloendotheliosis virus. Avian Dis. 140:289–312.
21:77–89. 233 Tsai, W.P, T.D Copeland, and S. Oroszlan. 1986.
220 Takagi, M., K. Ishikawa, H. Nagai, T. Sasaki, K. Gotoh, Biosynthesis and chemical and immunological
and H. Koyama. 1996. Detection of contamination of characterization of avian reticuloendotheliosis virus
vaccines with the reticuloendotheliosis virus by env gene‐encoded proteins. Virology. 155:567–583.
reverse transcriptase polymerase chain reaction 234 Tsai, W.P, and S. Oroszlan. 1988. Site‐directed
(RT‐PCR). Virus Res. 40:113–121. cytotoxic antibody against the C‐terminal segment of
221 Taniguchi, T., N. Yuasa, S. Sato, and T. Horiuchi. 1977. the surface glycoprotein gp90 of avian
Pathological changes in chickens inoculated with reticuloendotheliosis virus. Virology. 166:608–611.
reticuloendotheliosis‐virus‐contaminated Marek’s 235 Vogt, P.K, J.L Spencer, W. Okazaki, R.L Witter, and
disease vaccine. Natl Inst Anim Health Q (Tokyo). L.B Crittenden. 1977. Phenotypic mixing between
17:141–150. reticuloendotheliosis virus and avian sarcoma viruses.
222 Taylor, H.W, and L.D Olson. 1973. Chronologic study Virology. 80:127–135.
of the T‐virus in chicks. II. Development of 236 Wakabayashi, T., and H. Kawamura. 1975. Virus of
hematologic changes. Avian Dis. 17:794–802. reticuloendotheliosis virus group: Persistent infection
223 Temin, H.M, and V.K Kassner. 1974. Replication of in chickens and viral transmission to fertile egg. In:
reticuloendotheliosis viruses in cell culture: acute 79th Annual Meeting of the Japanese Society of
infection. J Virol. 13:291–297. Veterinay Science. pp 12–13.
224 Temin, H.M, and V.K Kassner. 1975. Replication of 237 Wakabayashi, T., and H. Kawamura. 1977. Serological
reticuloendotheliosis viruses in cell culture: chronic survey of reticuloendotheliosis virus infection among
infection. J Gen Virol. 27:267–274. chickens in Japan. Natl Inst Anim Health Q (Tokyo).
225 Temin, H.M, E. Keshet, and S.K Weller. 1980. 17:73–74.
Correlation of transient accumulation of linear un‐ 238 Wakenell, P.S, A.M Fadly, K.A Schat, and J.L Spencer.
integrated viral DNA and transient cell killing by avian 1997. An overview of problems in diagnosis of
leukosis and reticuloendotheliosis viruses. In: Cold neoplastic diseases of poultry. In: Diagnosis and
Spring Harbor Symposium on Quantitative Biology. Control of Neoplastic Diseases of Poultry. American
Cold Spring Harbor, NY. 773–778. Association of Avian Pathologists, Kennet Square, PA.
226 Terada, N., T. Kuramoto, and T. Ino. 1977. pp 1–5.
Comparison of susceptibility to the T strain of 239 Waldrip, D.W 1994. RE‐like syndrome. In: 29th
reticuloendotheliosis virus among families of Japanese National Meeting on Poultry Health and
quail. Japanese Poultry Science. 14:259–265. Condemnations. Salisbury, MD. 113–113.
2 27 Theilen, G.H, R.F Zeigel, and M.J Twiehaus. 1966. 240 Walker, M.H, B.J Rup, A.S Rubin, and H.R Bose, Jr.
Biological studies with RE virus (strain T) that 1983. Specificity in the immunosuppression induced
induces reticuloendotheliosis in turkeys, chickens, by avian reticuloendotheliosis virus. Infect Immun.
and Japanese quail. J Natl Cancer Inst. 40:225–235.
37:731–743. 241 Wang, X.L, Z. Zhang, S.J Jiang, and Z.Z Cui. 2005.
228 Thompson, K.D, R.G Fischer, and D.H Luecke. 1968. Immunogenicity of envelope glycoprotein gene of
Determination of the viremic period of avian reticuloendotheliosis virus expressed in insect cell.
reticuloendotheliosis virus (strain T) in chicks and Wei Sheng Wu Xue Bao. 45:593–597.
virus viability in Triatoma infestans (Klug) 242 Watanabe, S., and H.M Temin. 1982. Encapsidation
[Hemiptera: Reduviidae]. Avian Dis. 12:354–360. sequences for spleen necrosis virus, an avian
710 Section II  Viral Diseases

retrovirus, are between the 5′ long terminal repeat 257 Witter, R.L, H.G Purchase, and G.H Burgoyne. 1970.
and the start of the gag gene. Proc Natl Acad Sci USA. Peripheral nerve lesions similar to those of Marek’s
79:5986–5990. disease in chickens inoculated with
243 Watanabe, S., and H.M Temin. 1983. Construction of reticuloendotheliosis virus. J Natl Cancer Inst.
a helper cell line for avian reticuloendotheliosis virus 45:567–577.
cloning vectors. Mol Cell Biol. 3:2241–2249. 258 Witter, R.L, and D.W Salter. 1989. Vertical
244 Weaver, T.A, K.J Talbot, and A.T Panganiban. 1990. transmission of reticuloendotheliosis virus in breeder
Spleen necrosis virus gag polyprotein is necessary for turkeys. Avian Dis. 33:226–235.
particle assembly and release but not for proteolytic 259 Witter, R.L, J.M Sharma, and A.M Fadly. 1986.
processing. J Virol. 64:2642–2652. Nonbursal lymphomas induced by nondefective
245 Wei, K., Z. Sun, S. Zhu, W. Guo, P. Sheng, Z. Wang, C. reticuloendotheliosis virus. Avian Pathol. 15:467–486.
Zhao, Q. Zhao, and R. Zhu. 2012. Probable congenital 260 Witter, R.L, E.J Smith, and L.B Crittenden. 1981.
transmission of reticuloendotheliosis virus caused by Tolerance, viral shedding, and neoplasia in chickens
vaccination with contaminated vaccines. PLoS One. infected with non‐defective reticuloendotheliosis
7:e43422. viruses. Avian Dis. 25:374–394.
246 Weinstock, D., K.A Schat, and B.W Calnek. Cytotoxic 261 Wong, T.C, and M.M Lai. 1981. Avian
1989. T lymphocytes in reticuloendotheliosis virus‐ reticuloendotheliosis virus contains a new class of
infected chickens. Eur J Immunol. 19:267–272. oncogene of turkey origin. Virology. 111:289–293.
247 Wilhelmsen, K.C, K. Eggleton, and H.M Temin. 1984. 262 Yamada, S., S. Kamikawa, Y. Uchinuno, H. Fujikawa,
Nucleic acid sequences of the oncogene v‐rel in K. Takeuchi, A. Tominaga, and K. Matsura. 1977.
reticuloendotheliosis virus strain T and its cellular Distribution of antibody against reticuloendotheliosis
homolog, the proto‐oncogene c‐rel. J Virol. virus and isolation of the virus. J Jpn Vet Med Assoc.
52:172–182. 30:387–390.
248 Wilhelmsen, K.C, and H.M Temin. 1984. Structure 263 Yaniv, A., A. Gazit, M. Ianconescu, K. Perk, B.
and dimorphism of c‐rel (turkey), the cellular Aizenberg, and A. Zimber. 1979. Biochemical
homolog to the oncogene of reticuloendotheliosis characterization of the type C retrovirus associated
virus strain T. J Virol. 49:521–529. with lymphoproliferative disease of turkeys. J Virol.
249 Witter, R.L 1997. Avian tumor viruses: persistent and 30:351–357.
evolving pathogens. Acta Vet Hung. 45:251–266. 264 Yao, Y., D. Vasoya, L. Kgosana, L.P Smith, Y. Gao, X.
250 Witter, R.L 2006. Prevention and control of Wang, M. Watson, and V. Nair. 2017. Activation of
reticuloendotheliosis virus infection: rationale and gga‐miR‐155 by reticuloendotheliosis virus T strain
strategies. In: AAAP Avian Tumor Virus Symp. and its contribution to transformation. J Gen Virol.
Honolulu, HI. 81–89. 98:810–820.
251 Witter, R.L, and L.B Crittenden. 1979. Lymphomas 265 Yoshida, I., M. Sakata, K. Fujita, T. Noguchi, and N.
resembling lymphoid leukosis in chickens inoculated Yuasa. 1981. Modification of low virulent Newcastle
with reticuloendotheliosis virus. Int J Cancer. disease virus infection in chickens infected with
23:673–678. reticuloendotheliosis virus. Natl Inst Anim Health Q
252 Witter, R.L, and S.E Glass. 1984. Reticuloendotheliosis (Tokyo). 21:1–6.
in breeder turkeys. Avian Dis. 28:742–750. 266 Yuasa, N., I. Yoshida, and T. Taniguchi. 1976. Isolation
253 Witter, R.L, I.M Gimeno, and A.M Fadly 2005. of a reticuloendotheliosis virus from chickens
Differential diagnosis of lymphoid and myeloid inoculated with Marek’s disease vaccine. Natl Inst
tumors in the chicken. In: AAAP Slide Study Set #27 Anim Health Q (Tokyo). 16:141–151.
American Association of Avian Pathologists, Athens, 267 Zavala, G. 2006. Immunosuppression induced by
GA (Electronic media). 1–49. reticuloendotheliosis virus. In: Proc. AAAP Avian
254 Witter, R.L, and D.C Johnson. 1985. Epidemiology of Tumor Virus Symp. Honolulu, HI. 70–80.
reticuloendotheliosis virus in broiler breeder flocks. 268 Zavala, G., S. Cheng, T. Barbosa, and H. Haefele. 2006.
Avian Dis. 29:1140–1154. Enzootic reticuloendotheliosis in the endangered
255 Witter, R.L, L.F Lee, L.D Bacon, and E.J Smith. 1979. Attwater’s and greater prairie chickens. Avian Dis.
Depression of vaccinal immunity to Marek’s disease 50:520–525.
by infection with reticuloendotheliosis virus. Infect 269 Zavala, G., A.M Fadly, and H. Hunt. Oncornaviruses
Immun. 26:90–98. Leukosis/Sarcoma and Reticuloendotheliosis. In:
256 Witter, R.L, I.L Peterson, E.J Smith, and D.C Johnson. Isolation, Identification and 2016. Characterization of
1982. Serologic evidence in commercial chicken and Avian Pathogensd. S.M Williams, L. Dufour‐Zavala,
turkey flocks of infection with reticuloendotheliosis M.W Jackwood, M.D Lee, B. Lupiani, W.M Reed, E.
virus. Avian Dis. 26:753–762. Spackman and P.R Woolcock, eds. American
Chapter 15  Neoplastic Diseases 711

Association of Avian Pathologists, Athens, GA. 12 Ball, R. 1945. Two unusual neoplasms in the chicken
269–283. iris. Cornell Vet. 35:383–386.
270 Zeigel, R.F, G.H Theilen, and M.J Twiehaus. 1966. 13 Barua, A., P. Bitterman, J.S Abramowicz, A.L Dirks, J.M
Electron microscopic observations on RE virus (strain Bahr, D.B Hales, M.J Bradaric, S.L Edassery, J.
T) that induces reticuloendotheliosis in turkeys, Rotmensch, and J.L Luborsky. 2009. Histopathology of
chickens, and Japanese quail. J Natl Cancer Inst. ovarian tumors in laying hens: a preclinical model of
37:709–729. human ovarian cancer. Int J Gynecol Cancer. 19:531–539.
71 Zhai, S.L, S.N Chen, T. Lin, X.H Wen, W.K Wei, D.H
2 14 Beard, J.W, E.A Hillman, D. Beard, K. Lapis, and U.
Lv, and R.A Chen. 2016. Emergence of Heine. 1975. Neoplastic response of the avian liver to
reticuloendotheliosis virus in pigeons in Guangdong host infection with strain MC29 leukosis virus. Cancer
Province, Southern China. Arch Virol. 161:2007–2011. Res. 35:1603–1627.
72 Ziegel, R.F 1961. Morphological evidence of the
2 15 Beasley, J.N, S. Klopp, and B. Terry. 1986. Neoplasms in
association of virus particles with the pancreatic the oviducts of turkeys. Avian Dis. 30:433–437.
acinar cells of the chick. J Natl Cancer Inst. 26:1011–1039. 16 Beer, J.V S., G.W 1961. An ovarian tumour in a Mallard.
Bull Brit Ornithologists Club. 81:153–156.
17 Berry, W., A. Doernte, M. Conner, M. Barnes, and S.
Other Tumors
Oates. 2006. Spontaneously occurring fibroid tumors of
1 Abels, H. 1929. Die Geschwulste der Vogelhaut. the laying hen oviduct. Poult Sci. 85:1969–1974.
Z Krebsforsch. 29:207–210. 18 Bolte, A.L, and E. Burkhardt. 2000. A teratoma in a
2 Alfonso, M., L. Adochiles, V.M Hendrickson, D.K Muscovy duck (Cairina moschata). Avian Pathol.
Carver, G.C Rodriguez, and H.J Barnes. 2005. 29:237–239.
Metastatic adenocarcinoma in the lungs of older laying 19 Broman, M., J. Brown, G. Zavala, and K. Keel. 2011.
hens. Avian Dis. 49:430–432. Clinical Challenge. J Zoo Wildl Med. 42:766–769.
3 Anderson, W.I, P.C McCaskey, K.A Langheinrich, and 20 Campbell, J.G 1969. Tumours of the Fowl. Lippincott,
A.E Dreesen. 1985. Neurofibrosarcoma and leiomyosar­ Philadelphia, Pennsylvania. 1–292.
coma in slaughterhouse broilers. Avian Dis. 33:827–828. 21 Campbell, J.G, and E.C Appleby. 1966. Tumours in
4 Anjum, A.D 1987. Adenocarcinoma of the oviduct of young chickens bred for rapid body growth (broiler
the domestic fowl (Gallus domesticus) and its chickens): A study of 351 cases. J Pathol Bacteriol.
relationship to steriod sex hormones. London, United 92:77–90.
Kingdom, Royal Veterinary College. PhD: 1–356. 22 Cardona, C.J, A.A Bickford, and K. Emanuelson. 1992.
5 Anjum, A.D, and L.N Payne. 1988. Concentration of Squamous cell carcinoma on the legs of an Aracauna
steriod sex hormones in the plasma of hens in relation chicken. Avian Dis. 36:474–479.
to oviduct tumours. Br Poult Sci. 29:729–734. 23 Carnaghan, R.B A. 1965. Hepatic tumours in ducks fed
6 Anjum, A.D, L.N Payne, and E.C Appleby. 1988. a low level of toxic groundnut meal. Nature. (Lond).
Spontaneous occurrence and experimental induction of 208:308.
leiomyoma of the ventral ligament of the oviduct of the 24 Childs‐Sanford, S.E, K.M Rassnick, and A. Alcaraz.
hen. Res Vet Sci. 45:341–348. 2006. Carboplatin for treatment of a Sertoli cell tumor
7 Ansenberger, K., C. Richards, Y. Zhuge, A. Barua, J.M in a mallard (Anas platyrhynchos). Vet Comp Oncol.
Bahr, J.L Luborsky, and D.B Hales. 2010. Decreased 4:51–56.
severity of ovarian cancer and increased survival in 25 Christopher, J., P. Rama Rao, J.V Narayana, and G.A
hens fed a flaxseed‐enriched diet for 1 year. Gynecol Sastry. 1965. A case of leiomyoma in the liver of a
Oncol. 117:341–347. pigeon. Indian Vet J. 42:754–755.
8 Apperly, F.L 1935. Primary carcinoma of the lung in the 26 Cole, R.K 1946. An avian retinoblastoma. Cornell Vet.
domestic fowl. Am J Cancer. 23:556–557. 36:350–353.
9 Arshad, S.S, A.P Bland, S.M Hacker, and L.N Payne. 27 Couvillion, C.E, W.A Maslin, and R.M Montgomery.
1997. A low incidence of histiocytic sarcomatosis 1990. Multiple feather follicle cysts in a wild turkey.
associated with infection of chickens with the J Wildl Dis. 26:122–124.
HPRS‐103 strain of subgroup J avian leukosis virus. 28 Desserich, M., D.W Folsch, and V. Ziswiler. 1984. Das
Avian Dis. 41:947–956. Schnabelkupieren bei Huhnern. Tier Praxis.
10 Baker, J.R 1980. A proventricular adenoma in a 12:191–202.
Brazilian teal (Amazonetta brasiliensis). Vet Rec. 29 Dinev, I. 2013. Enzootic outbreak and spontaneous
107:63–64. regression of keratoacanthomas in a commercial layer
11 Balaquer, L., J. Romano, and A. Mora. 1995. A poorly‐ flock. Acta Vet Hung. 61:366–375.
differentiated squamous cell thymoma in a chicken 30 Dittmer, K.E, A.F French, D.J Thompson, K.N Buckle,
with lymphoma. Avian Pathol. 24:737–741. and K.G Thompson. 2012. Primary bone tumors in
712 Section II  Viral Diseases

birds: A review and description of two new cases. 46 Gorham, S.L, and M.A Ottinger. 1986. Sertoli cell
Avian Dis. 56:422–426. tumors in Japanese quail. Avian Dis. 30:337–339.
31 Duflot, A., R. Mehrotra, S.‐Z. Yu, L. Barraud, C. Trepo, 47 Grewel, G.S, and H.S Patel. 1985. An unusual case of
and L. Cova. 1995. Spectrum of liver disease and Duck rhabdomyosarcoma in a fowl. Avian Pathol. 14:177–180.
Hepatitis B virus infection in a large series of Chinese 48 Guerin, M. 1954. Tumeurs spontanees de la poule. In:
ducks with hepatocellular carcinoma. Hepatology. Tumeurs Spontanees des Animaux de Laboratoire
21:1483–1491. Legrand, Paris, France. 153–180.
32 Dukes, T.W, and J.R Pettit. 1983. Avian ocular 49 Guthrie, J. 1967. Specificity of the metallic ion in the
neoplasia—a description of spontaneously occurring experimental induction of teratomas in fowl. Br J
cases. Can J Comp Med. 47:33–36. Cancer. 21:619–622.
33 Fallavena, L.C, C.W Canal, C.T Salle, H.L Moraes, S.L 50 Hafner, S. 2011. Unpublished observations.
Rocha, R.A Pereira, and A.B da Silva. 2002. Presence of 51 Hafner, S., M.A Goodwin, E.J Smith, D.I Bounous, M.
avipoxvirus DNA in avian dermal squamous cell Puette, L.C Kelley, K.A Langheinrich, and A.M Fadly.
carcinoma. Avian Pathol. 31:241–246. 1996. Multicentric histiocytosis in young chickens.
34 Fernandez‐Bellon, H., J. Martorell, R. Rabanal, and A. Gross and light microscopic pathology. Avian Dis.
Ramis. 2003. Rhabdomyosarcoma in a racing pigeon 40:202–209.
(Columba livia). Avian Pathol. 32:613–616. 52 Hafner, S., B.G Harmon, G.N Rowland, R.G Stewart,
35 Foster, D.N, K.E Nestor, Y.M Saif, W.L Bacon, and P.D and J.R Glisson. 1991. Spontaneous regression of
Moorhead. 1989. Influence of selection for increased “dermal squamous‐cell carcinoma” in young chickens.
body weight on the incidence of leiomyomas and Avian Dis. 35:321–327.
leiomyosarcomas in Japanese quail. Poult Sci. 53 Hafner, S., B.G Harmon, R.G Stewart, and G.N
68:1447–1453. Rowland. 1993. Avian keratoacanthoma (dermal
36 Frankenhuis, M.T 1987. Sex reversal in poultry. Poultry squamous cell carcinoma) in broiler chicken carcasses.,
(Misset). 32:46–47. Vet Pathol. 30:265–270.
37 Fredrickson, T.N 1987. Ovarian tumors of the hen. 54 Hafner, S., and K. Latimer. 1997. Cutaneous mast cell
Environ Health Perspect. 73:35–51. tumours with pulmonary metastasis in a hen. Avian
38 Fredrickson, T.N, and C.F Helmboldt. 1991. Tumors of Pathol. 26:657–663.
unknown etiology. In: Diseases of Poultry, 9th ed, B.W. 55 Hafner, S., K. Latimer, L.C Kelley, K. Wortham, and M.
Calnek, H.J. Barnes, C.W. Beard, W.M. Reid and H.W. Puette. 1995. Malignant melanocytic neural crest
Yoder Jr, eds. Iowa State University Press, Ames, Iowa. tumor in a young chicken (Gallus domesticus). Vet
459–470. Pathol. 32:535–537.
39 Gentle, M.J 1986. Neuroma formation following partial 56 Hafner, S., R.L Reece, and S.M Williams. 2013. Other
beak amputation (beak trimming) in the chicken. Res tumors. In: Diseases of Poultry, 13th, D.E. Swayne, J.R.
Vet Sci. 41:383–385. Glisson, L.R. McDougald, L.K. Nolan, D.L. Suarez and
40 Gentle, M.J, and L.H Hunter. 1988. Neural V. Nair, eds. Wiley‐Blackwell, Ames, Iowa. 604–673.
consequences of partial toe amputation in chickens. Res 57 Hakim, A.A, C.P Barry, H.J Barnes, K.E Anderson, J.
Vet Sci. 45:374–376. Petitte, R. Whitaker, J.M Lancaster, R.M Wenham, D.K
41 Gentle, M.J, B.H Thorp, and B.O Hughes. 1995. Carver, J. Turbov, A. Berchuck, L. Kopelovich, and G.C
Anatomical consequences of partial beak amputation Rodriguez. 2009. Ovarian adenocarcinomas in the
(beak trimming) in turkeys. Res Vet Sci. 58:158–162. laying hen and women share similar alterations in p53,
42 Giles, J.R, H.L Shivaprasad, and P.A Johnson. 2004. ras, and HER‐2/neu. Cancer Prev Res (Phila). 2:114–121.
Ovarian tumor expression of an oviductal protein in 58 Hall, S.M, D. Mosier, and M.M Degraw. 1994.
the hen: a model for human serous ovarian Histological and ultrastructural characterization of a
adenocarcinoma. Gynecol Oncol. 95:530–533. cutaneous mast cell tumor in a chicken. In: 43rd
43 Good, R.E 1991. The importance of squamous cell Western Poultry Disease Conference, University of
carcinoma in broilers. In: Avian Tumor Virus California, Davis, California. 6–7.
Symposium. American Association of Avian 59 Haritani, M., H. Kajigaya, T. Akashi, M. Kamemura, N.
Pathologists. 56–57. Tanahara, M. Umeda, M. Sugiyama, M. Isoda, and C.
44 Goodchild, W.M 1969. Adenocarcinoma of the oviduct Kato. 1984. A study on the origin of adenocarcinoma in
in laying hens. Vet Rec. 84:122. fowls using immunohistological technique. Avian Dis.
45 Goodwin, M.A, S. Hafner, D.I Bounous, J. Brown, E. 28:1130–1134.
Smith, and A. Fadly. 1999. Multicentric histiocytosis: 60 Harris, E.A, O.J Fletcher, K.E Anderson, J. Petitte, L.
experimental induction in broiler and specific Kopelovich, and P.E Mozdziak. 2014. Epithelial cell
pathogen‐free leghorn chickens. Avian Pathol. tumors of the hen reproductive tract. Avian Dis.
28:273–278. 58:95–101.
Chapter 15  Neoplastic Diseases 713

61 Hatai, H., K. Ochiai, and T. Umemura. 2009. Detection 77 Manarolla, G., S. Caserio, G. Sironi, and T. Rampin.
of avian leukosis virus genome by a nested polymerase 2011. Morphological and immunohistochemical
chain reaction using DNA and RNA from dried feather observations on leiomyoma of the ventral ligament of
shafts. J Vet Diagn Invest. 21:519–522. the oviduct of the hen. J Comp Pathol. 144:180–186.
62 Helmboldt, C.F, G. Migaki, K.A Langheinrich, and R.M 78 Mawdesley‐Thomas, L.E, and D.H Solden. 1967.
Jakowski. 1974. Teratoma in domestic fowl (Gallus Osteogenic sarcoma in a domestic goose (Anser anser).
gallus). Avian Dis. 18:142–148. Avian Dis. 11:365–370.
63 Himmel, L., and R. Cianciolo. 2017. Nodular 79 Menezes, R.C, R. Tortelly, D.C Gomes, and R.M Pinto.
typhlocolitis, heterakiasis, and mesenchymal neoplasia 2003. Nodular typhlitis associated with the nematodes
in a ring‐necked pheasant (Phasianus colchicus) with Heterakis gallinarum and Heterakis isolonche in
immunohistochemical characterization of visceral pheasants: frequency and pathology with evidence of
metastases. J Vet Diagn Invest. DOI:1040638717707555. neoplasia. Mem Inst Oswaldo Cruz. 98:1011–1016.
64 Ilchmann, G., and V. Bergmann. 1975. Histologische 80 Movassaghi, A.R, and K. Sardari. 2008. Cutaneous
and elektronenmikroskopische untersuchungen zu leiomyosarcoma in a pigeon. Vet Rec. 162:250–251.
adenokarzinomatose der legehennen. Arch Exp 81 Nakamura, K., M. Ito, K. Fujino, Y. Yamamoto, M.
Veterinaermed. 29:897–907. Mase, M. Yamada, H. Kobayashi, and T. Harada. 2010.
65 Jackson, C. 1936. The incidence and pathology of Pathology and microbiology of dermal squamous cell
tumors of domesticated animals in South Africa. carcinoma in young brown chickens reared on reused
Onderstepoort J Vet Res. 6:1–460. litter. Avian Dis. 54:1120–1124.
66 James, C. 1968. Neoplasms of the chicken. Ceylon Vet J. 82 Newman, S.J, and K. West. 2001. Pulmonary
16:59–61. leiomyosarcoma in a white Carneau pigeon (Columba
67 Kajigaya, H., M. Kamemura, N. Tanahara, A. Ohta, H. livia). J Avian Med Surg. 15:293–296.
Suzuki, M. Sugiyama, and M. Isoda. 1987. The 83 Nili, H., and W.R Kelly. 1996. Form and function of
influence of celomic membranes and a tunnel between lacunae in the ovary of the laying hen. Anat Rec. 244:165–174.
celomic cavities on cancer metastasis in poultry. Avian 84 Nobel, T.A, F. Neumann, and Dison. 1964. A
Dis. 31:176–186. histological study of peritoneal carcinomatosis in the
68 Kawaguchi, T., K. Nomura, Y. Hirayama, and T. laying hen. Avian Dis. 8:513–522.
Kitagawa. 1987. Establishment and characterization of 85 Ochiai, K., K. Ohashi, T. Mukai, T. Kimura, T.
a chicken hepatocellular carcinoma cell line, LMH. Umemura, and C. Itakura. 1999. Evidence of neoplastic
Cancer Res. 47:4460–4464. nature and viral aetiology of so‐called fowl glioma. Vet
69 Kelley, L., J. Hill, S. Hafner, and K. Langheinrich. 1993. Rec. 145:79–81.
Enterogenous cysts in chickens. Vet Pathol. 30:376–378. 86 Okoye, J.O, and C.C Ilochi. 1993. Pancreatic
70 Kheirandish, R., S. Azizi, M. Salehi, S. Shojaeepour, and adenocarcinoma in guinea fowl. Avian Pathol. 22:401–406.
S. Hassanzadeh. 2016. Metastatic leiomyosarcoma 87 Olson, C., and K.L Bullis. 1942. A survey of
originating in the thigh skeletal muscle of a Larry breed spontaneous neoplastic diseases in chickens.
hen. J Avian Med Surg. 30:141–145. Massachusetts Agric Exp Stat Bull. 391:1–25.
71 Krogh, G. 1953. Two cases of rhabdomyosarcoma in 88 Ottinger, M.A, E. Adkinsregan, J. Buntin, M.F Cheng,
chickens. Nord Vet Med. 5:232–236. T. Devoogd, C. Harding, and H. Opel. 1984. Hormonal
72 Kusewitt, D.F, R.L Reece, and K.B Miska. 1997. S‐100 mediation of reproductive behavior. J Exp Zool. 232:605–616.
immunoreactivity in melanomas of two marsupials, a 89 Palmieri, C., M. Romanucci, P. Loi, and L. Della Salda.
bird, and a reptile. Vet Pathol. 34:615–618. 2012. Intracoelomic teratoma in a domestic duck (Anas
73 Leach, S., J.J Heatley, R.R Pool, Jr., and K. Spaulding. platyrhynchos domesticus): A case report including
2008. Bilateral testicular germ cell‐sex cord‐stromal immunohistochemistry and electron microscopy. Res
tumor in a pekin duck (Anas platyrhynchos Vet Sci. 93:862–864.
domesticus). J Avian Med Surg. 22:315–319. 90 Pandiri, A.R, I.M Gimeno, W.M Reed, S.D Fitzgerald,
74 Ling, Y.S, and Y. Guo. 1985. Pathological study of and A.M Fadly. 2009. Subgroup J avian leukosis virus‐
spontaneous mesothelioma in ducks. Chin J Vet Sci. induced histiocytic sarcomatosis occurs only in
Technol 9:15–16. persistenly viremic but not immunotolerized meat‐type
75 Lombard, L.S, and E.J Witte. 1959. Frequency and types chickens. Vet Pathol. 46:282–287.
of tumors in mammals and birds of the Philadelphia 91 Parnaik, G.M, and D. Mohanty. 1970. A case of avian
Zoological Garden. Cancer Res. 19:127–141. mastocytoma. Indian Vet J. 47:298–300.
76 Madekurozwa, M.C, and W.H Kimaro. 2008. An 92 Payne, L.N, A.M Gillespie, and K. Howes. 1992.
ultrastructural characterization of the ooplasm in Myeloid leukaeomogenicity and transmission of the
ovarian follicles of the immature ostrich (Struthio HPRS‐103 strain of avian leukosis virus. Leukaemia.
camelus). Anat Histol Embryol. 37:214–218. 6:1167–1176.
714 Section II  Viral Diseases

93 Paździor, K., M. Szweda, I. Otrocka‐Domagała, and T. 111 Siller, W.G 1956. A Sertoli cell tumour causing
Rotkiewicz. 2012. Extragonadal teratoma in a feminization in a brown leghorn capon. J Endocrinol.
domestic turkey (Meleagris gallopavo domestica). 14:197–203.
Avian Pathol. 41:285–289. 112 Sokkar, S.M, M.A Mohammed, A.J Zubaidy, and A.
94 Porter, T.E, B.M Hargis, J.L Silsby, and M.E El‐ Mutalib. 1979. Study of some non‐leukotic avian
Halawani. 1989. Differential steroid production neoplasms. Avian Pathol. 8:69–75.
between theca interna and theca externa cells: A three 113 Stewart, H.L 1966. Pulmonary cancer and
cell model for follicular steroidogenesis in avian adenomatosis in captive wild mammals and birds
species. Endocrinology. 125:109–116. from the Philadelphia Zoo. J Natl Cancer Inst.
95 Purvulov, B., and S. Bozhkov. 1984. Pathology of some 36:117–138.
spontaneous neoplasms of fowls. Obshch i 114 Sugiyama, M., H. Yamashina, T. Kanbara, H. Kajigaya,
Stravnitelna Patologiya. 16:55–58. K. Konagaya, M. Umeda, M. Isoda, and T. Sakai. 1987.
96 Razmyar, J., O. Dezfoulian, B. Shojadoost, M. Dermal squamous cell carcinoma in a laying hen. Jpn J
Masoudifard, and S.M Peighambari. 2005. Sertoli cell Vet Sci. 49:1129–1130.
tumor in a pigeon (Columba livia). J Avian Med Surg. 115 Swayne, D.E, G.N Rowland, and O.J Fletcher. 1986.
19:286–288. Pinealoma in a broiler breeder. Avian Dis. 30:853–855.
97 Reece, R.L 1992. Observations on naturally occurring 116 Tomioka, Y., K. Ochiai, K. Ohashi, T. Kimura, and T.
neoplasms in birds in the state of Victoria, Australia. Umemura. 2003. In ovo infection with an avian
Avian Pathol. 21:3–32. leukosis virus causing fowl glioma: viral distribution
98 Reece, R.L 2008. Other tumors of unknown etiology. and pathogenesis. Avian Pathol. 32:617–624.
In: Diseases of Poultry, 12th, Y.M. Saif, A.M. Fadly, 117 Turnquest, R.U 1979. Dermal squamous cell
J.R. Glisson, L.R. McDougald, L.K. Nolan and D.E. carcinoma in young chickens. Am J Vet Res. 40:1628–1633.
Swayne, eds. Blackwell Publishing, Ames, Iowa. 118 Uetsuka, K., T. Suzuki, K. Doi, and T. Nunoya. 2012.
593–616. Malignant Sertoli Cell tumor in a goose (Anser
99 Reece, R.L 1996. Some observations on naturally cygnoides domesticus). Avian Dis. 2012:781–785.
occurring neoplasms in domestic fowl in the state of 119 Walser, M.M, and P.S Paul. 1979. Ovarian
Victoria, Australia. Avian Pathol. 25:407–447. adenocarcinomas in domestic turkeys. Avian Pathol.
100 Reece, R.L 1995. Unpublished observations. 8:335–339.
101 Reece, R.L 2011. Unpublished observations. 120 Wang, Z., L. Tang, G. Sun, Y. Tang, Y. Xie, S. Wang, X.
102 Reece, R.L 2001. Unpublished observations. Hu, W. Gao, S.B Cox, and J.S Wang. 2006. Etiologic
103 Reece, R.L, and S.A Lister. 1993. An abdominal study of esophageal squamous cell carcinoma in an
teratoma in a domestic goose (Anseriformes, Anser endemic region: a population‐based case control
anser domesticus). Avian Pathol. 22:193–196. study in Huaian, China. BMC Cancer. 6:287.
104 Rigdon, R.H 1970. Spontaneous‐occurring tumors in 121 Warner, N.E, N.B Friedman, B.J Bomze, and F. Masin.
the duck: Review of the literature and report of three 1960. Comparative pathology of experimental and
cases. Avian Dis. 14:431–444. spontaneous androblastomas and gynoblastomas of
105 Rubio, C.A, and F.S Liu. 1989. Spontaneous squamous the gonads. Am J Obstet Gynecol. 79:971–988.
carcinoma of the esophagus in chickens. Cancer. 122 Weinstock, D., M.T Correa, D.V Rives, and D.P
64:2511–2514. Wages. 1995. Histopathology and epidemiology of
106 Sanger, V.L, and A. Lagace. 1966. Avian condemnations due to squamous cell carcinoma in
xanthomatosis. Etiology and pathogenesis. Avian Dis. broiler chickens in North Carolina. Avian Dis.
10:103–113. 39:676–686.
107 Saunders, L.Z, and C.N Barron. 1958. Primary 123 Wight, P.A L. 1962. Gonadal maldevelopment in a
pigmented intraocular tumours in animals. Cancer flock of Rhode Island red fowls. J Endocrinol.
Res. 18:234–245. 23:341–349.
108 She, R.P 1987. Epidemiology and pathology of 124 Wight, P.A L. 1965. Neoplastic sequelae of gonadal
oropharyngo‐esophageal carcinoma in chickens from maldevelopment in a flock of domestic fowls. Avian
different areas in Zhongxian county, Hubei province. Dis. 9:327–335.
Acta Vet Zootech Sinica. 18:195–200. 125 Wight, P.A L., and J.G Campbell. 1976. Three unusual
109 Sievert, R. 2002. Pathomophologische intracranial tumours of the domesticated fowl. Avian
Untersuchungen zur Charakterisierung der Pathol. 5:201–214.
Hautkarzinomatose (Keratoakanthom) von 126 Williams, S.M, G. Zavala, S. Hafner, S.R Collett, and S.
Jungmasthühnern. Berlin, Freien Universität. PhD. Cheng. 2012. Metastatic melanomas in young broiler
110 Siller, W.G 1959. An osteogenic sarcoma in the fowl. chickens (Gallus gallus domesticus). Vet Pathol.
Br J Cancer. 13:642–646. 49:288–291.
Chapter 15  Neoplastic Diseases 715

127 Yokosuka, O., M. Omata, Y.Z Zhou, F. Imazeki, and K. 129 Zheng, S., L. Vuitton, I. Sheyhidin, V.D A., Y. Zhang
Okuda. 1985. Duck hepatitis B virus DNA in liver and and X. Lu. 2010. Northwestern China: a place to learn
serum of Chinese ducks: integration of viral DNA in a more on oesophageal cancer. Part one: behavioural
hepatocellular carcinoma. Proc Natl Acad Sci USA. and environmental risk factors. Eur J Gastroenterol
82:5180–5184. Hepatol. 22:917–925.
28 Zhang, J.L, F.C Liang, and Y.J Chen. 1985. Primary
1
pulmonary tumours in Pekin ducks: Pathological
analysis of 16 cases. Chin J Vet, Sci Technol. 4:32–33.

You might also like