You are on page 1of 8

Current Status and Future Directions of

Treatment Deintensification in Human


Papilloma Virus-associated Oropharyngeal
Squamous Cell Carcinoma
Bhishamjit S. Chera, MD,*,† and Robert J. Amdur, MD‡,§

The prevalence of patients with human papilloma virus (HPV)-associated oropharyngeal


squamous cell carcinoma (OPSCC) is rapidly increasing, and it is now well known that these
patients have a significantly better prognosis than patients with HPV-negative OPSCC.
Though standard treatments result in excellent cancer control, they are also associated with
substantial long-term toxicity. There is now great interest in evaluating less intensive (ie,
deintensified) treatment regimens to improve the therapeutic ratio (maintain excellent cancer
control and decrease toxicity). There are many different approaches that are being studied, and
each have their own caveats, with varying degrees of actual deintensification. In this article, we
critically review the current landscape of emerging deintensified treatment paradigms and
future direction of the treatment of HPV-associated OPSCC.
Semin Radiat Oncol 28:27-34 C 2017 Elsevier Inc. All rights reserved.

Introduction stratification based on HPV status, tobacco smoking history,


and nodal stage is now standardly used in clinical practice

T he prevalence of oropharyngeal squamous cell carcinoma


(OPSCC) has been rapidly increasing over the past 20-30
years and is thought to be directly related to the corresponding
when counseling patients on their prognosis. Patients with the
most favorable prognosis are those with HPV-associated
OPSCC, ≤ 10 pack years, and oN2c/N3 stage.6,8
increasing incidence of high-risk human papilloma virus There are 3 standard definitive treatment paradigms for
(HPV) infection of the oropharynx.1-3 Retrospective analyses OPSCC: (1) transoral surgery followed by pathological risk-
of several clinical trials have shown that patients with HPV- adapted radiotherapy (RT) and chemotherapy, (2) concurrent
associated OPSCC have a better response rate to chemotherapy RT and chemotherapy (ie, chemoradiotherapy [CRT]), and (3)
and radiation and a significantly better local-regional control neoadjuvant chemotherapy followed by CRT. The “preferred”
and overall survival (OS) with standard treatments as standard treatment depends on institutional biases; however,
compared to patients with HPV-negative tumors.4-7 Risk overall the most used organ preservation approach is con-
current CRT (without surgery and neoadjuvant chemo).
*Department of Radiation Oncology, University of North Carolina School of These standard treatments result in excellent cancer control
Medicine, Chapel Hill, NC. and survival in patients with HPV-associated OPSCC; how-

Lineberger Comprehensive Cancer Center, University of North Carolina ever, they are associated with substantial toxicity.9-13 Thus, we

Hospitals, Chapel Hill, NC. may be “over treating” many patients with HPV-associated
Department of Radiation Oncology, University of Florida Hospitals,
Gainesville, FL.
OPSCC. There is now great interest in reducing the intensity of
§
Shands Cancer Center, University of Florida Hospitals, Gainesville, FL. treatment with the goal of decreasing toxicity while maintain-
Funding source: University of North Carolina School of Medicine, Department ing cancer control (ie, improving the therapeutic ratio). There
of Radiation Oncology and University of Florida School of Medicine, are many ways to deintensify treatment each with their own
Department of Radiation Oncology. “pros” and “cons,” and often the reduction in treatment
Conflicts of interest: none.
Address reprint requests to Bhishamjit S. Chera, MD, Department of Radiation
intensity is small or reduction in 1 treatment modality is
Oncology, University of North Carolina Hospitals, 101 Manning Dr, CB offset with an increase in another.14 We here-in review and
#7512, Chapel Hill, NC 27599-7512. E-mail: bchera@med.unc.edu discuss the various ongoing deintensification approaches and

http://dx.doi.org/10.1016/j.semradonc.2017.08.001 27
1053-4296/& 2017 Elsevier Inc. All rights reserved.
28 B.S. Chera and R.J. Amdur

forecast the future direction of deintensification for HPV- was 94% and the 5-year local-regional control for those with
associated OPSCC. and without RT indications was 84% vs 74%. However, in a
multicenter TLM study, patients who did not receive adjuvant
RT had worse overall and disease-free survival.16 One may
argue that all patients with node-positive OPSCC require
Transoral Surgery Paradigm adjuvant RT because of the risk of occult disease in the
Performing surgery first provides pathological information that ipsilateral retropharyngeal (~20%)20 and cranial level II nodes
can be used to tailor the use of adjuvant treatment, with the that are not surgically treated. Even in those who are node
goal of reducing or omitting radiation and chemotherapy. negative, the retropharyngeal nodes may still be at risk.
Proponents of the definitive surgery approach believe that the What about excluding the primary site from the RT treat-
toxicities of surgery ± radiation and chemotherapy are fewer ment volume? Many patients do not have indications for
than definitive CRT. Conventional surgery for OPSCC has postop RT related to the primary site (margin not close and no
been open en bloc resection with free flap reconstruction perineural invasion). In patients with indications for RT to the
which, when compared with CRT, has similar outcomes but neck (multiple positive nodes or extranodal tumor extension)
higher severe or fatal complications.13 but no indications at the primary site, it is reasonable to
Newer transoral surgical approaches (ie, transoral laser exclude the primary site operative bed from the RT target
microsurgery [TLM] and transoral robotic surgery [TORS]) volume.
are garnering much interest for the treatment of HPV- Historically, a negative margin was considered to be
associated OPSCC because these techniques are less invasive ≥5 mm, close margins were o5 mm, and positive was tumor
and thus have less toxicity. In fact, the toxicity of TLM and on the inked en bloc specimen. The definition of pathologic
TORS ± CRT or RT has been suggested to be less than margins is different with transoral surgical procedure. There is
definitive CRT.15,16 Because of the use of magnification and the no close margin designation and a negative margin is defined
transection of the tumor during surgery (TORS allows for en as a negative inked margin, regardless of distance.21 Margins
bloc resection), the “host-tumor” interface is better visualized, are defined and interpreted differently with transoral surgeries
allowing for more precise negative margin resections, max- because margins are carefully mapped intraoperatively under
imizing tumor removal, and limiting removal of normal better visualization (as compared to conventional surgical
healthy tissue, resulting in “surgically targeted therapy.16,17” techniques). Furthermore, for resection of the tonsil, the
Furthermore, similar to CRT, HPV-associated OPSCC is also superior constrictor muscle is taken with the resection and is
associated with a better prognosis when treated with primary considered a natural barrier of spread. The mean superior
surgery.7 The major potential benefits of primary TLM and constrictor thickness is ~2 mm, and even though tumor often
TORS are reduction in surgical morbidity and reduction in the abuts this muscle, because it is considered a barrier to spread,
intensity of CRT without compromising oncologic outcomes. what historically would be considered a close margin is
Primary surgery provides pathologic information that influen- considered negative with transoral surgery. Thus, close mar-
ces adjuvant treatment recommendations. RT and chemo- gins are not defined with transoral surgeries and the reported
therapy may be omitted or at least the RT dose may be reduced. rate of positive margins is low o10%, and therefore one could
The deintensification of RT and chemotherapy after TLM or argue that the primary site need not be treated with RT.
TORS has been arbitrary, institution specific, and has not been Omission of the primary site in intensity-modulated radio-
studied in a controlled manner. The eastern cooperative therapy (IMRT) plans will reduce the mean dose to the primary
oncology group (ECOG) is currently conducting a prospective site to ~40 Gy.22 Mean doses to the contralateral parotid and
randomized phase II study (ECOG3311) in which patients pharyngeal constrictors are not significantly reduced; however,
with HPV-associated T1-2, N1-N2b OPSCC all have transoral the mean dose to the oral cavity is reduced by a mean of
surgery followed by risk-adapted adjuvant radiation and 20 Gy.22 With primary site avoidance IMRT plans, swallowing
chemotherapy.18 function may not be improved. From the prospective series
from Haughey et al, patients with swallowing function scores
of 0-2 (normal swallowing to mild dysphagia) were not
Omission of Radiation significantly different between patients treated with TLM alone
There is no question that single modality treatment with TLM (47/52 patients) or TLM plus adjuvant RT (103/117); P ¼
and TORS alone will significantly reduce toxicity. Interestingly, 0.79. Thus, even the complete omission of RT did not improve
most patients (70%-80%) who receive TLM and TORS receive swallowing function. This suggests that sparing the primary
adjuvant RT.16 Currently on the ECOG3311, only ~11% of site with IMRT may not result in less dysphagia.
enrolled patients are being observed after transoral surgery (ie, The low-dose radiation bath associated with IMRT techni-
not receiving postoperative radiation and chemotherapy).18 ques makes it difficult to completely avoid or spare the primary
Thus, omission of RT may be a reality for only a small number site. Proton RT (especially intensity-modulated proton
of patients. therapy) has the advantage of eliminating the low-dose bath
Grant et al19 reported on 69 patients treated with TLM alone resulting in better avoidance of the primary site. The Mayo
at the Mayo Clinic; 44 patients who did not have indications clinic is currently conducting an observational study of patients
for RT and 25 patients who did have indications for RT in the with OPSCC who have indications for RT only to the neck after
neck, but refused. The 5-year local control for the entire group transoral resection, and neck dissection will be treated with
Treatment deintensification in HPV-associated OPSCC carcinoma 29

postoperative mucosal-sparing proton beam therapy ECS does not adversely affect disease control and survival in
(NCT02736786, clinicaltrials.gov). patients with HPV-associated OPSCC.28 Adjuvant chemo-
therapy may not be needed because of the enhanced radio-
curability of HPV-associated OPSCC.
Lowering the Radiation Dose
Transoral surgery will, at the very least, allow for a reduction in
RT dose. For patients with indications for RT, the standard Morbidity of Neck Dissection
adjuvant RT doses have been 60 Gy (close margins, ≥2 The reported long-term outcomes of transoral surgery studies
positive nodes, perineural invasion, and lymphovascular space focus on reporting excellent overall quality of life, preservation
invasion) or 66 Gy (positive margins or extracapsular spread of swallowing function, and low rates of permanent feeding
[ECS]). Thus, on average, patients with indications for RT tube use. Neck dissections are standardly part of the transoral
would be spared 4-10 Gy. It is debatable whether a TLM or surgical paradigm, which have associated known moderate
TORS procedure is less intensive than 10 Gy. Proponents of rates of morbidity, especially with the addition of postoperative
transoral surgery suggest that a 10-Gy reduction in dose to the RT. Many patients experience lymphedema, fibrosis, and
pharyngeal constrictors will have a significant effect on long- worsening cosemsis after a neck dissection and postoperative
term dysphagia.16 The available data (ie, definitive CRT) for the RT. Marginal mandibular and cranial nerve eleven (among
dose-effect relationship for dysphagia suggest that doses other nerves) injuries can occur as well. Proponents of transoral
≥50 Gy to the pharyngeal constrictors correlate with late surgery paradigm do not discuss or comment on the morbidity
dysphagia.23,24 The dose-response may be different in patients of neck dissections. Historically, avoidance of neck dissections
undergoing transoral surgery. In general, though, partial was thought to be important; however, the side effects of neck
sparing of pharyngeal constrictors should be more feasible dissection are not considered in the transoral surgery
when treating the oropharynx to a total dose of 60 vs 70 Gy. approach.
It is unknown whether lower adjuvant RT doses
(ie, ≤60 Gy) may be equally efficacious in HPV-associated
OPSCC. Ma and colleagues at the Mayo Clinic (Rochester, Neoadjuvant Chemotherapy
MN) have recently completed a phase II study evaluating a
de-escalated adjuvant therapy regimen of 30 Gy (1.5 Gy/fx
Paradigm
twice a day; intermediate pathological risk) or 36 Gy HPV-associated OPSCC has been observed to have a better
(1.8 Gy/fx, twice a day; high pathological risk) with concurrent response rate to neoadjuvant chemotherapy4 compared to
weekly docetaxel 15 mg/m2. Preliminary results (verbal com- HPV-negative disease, thus reducing the radiation dose
munication with Ma) show excellent local-regional control and and volume after which neoadjuvant chemotherapy may
preservation of swallowing function. They are currently be possible. The trade-off is an additional 9 weeks of
conducting a phase III study comparing this experimental intensive chemotherapy for a reduction of 1 week (ie,
arm to standard of care 60 Gy (2 Gy/fx, 4 times a day) with 10 Gy) of RT. A disadvantage of this approach is that the
concurrently weekly cisplatin 40 mg/m2 (NCT02908477, overall treatment time is increased, thus prolonging the
clinicaltrials.gov). The primary endpoint is grade 3-5 toxicity. duration, frequency, and severity of toxicities. The net
deintensification may be “zero” because 1 modality (ie, RT)
is being minimally decreased, while the other (ie, chemo-
Omission of Chemotherapy therapy) is maximally intensified.
Based on the individual Radiation Therapy Oncology Group Although the local-regional control of HPV-associated
(RTOG) and European Organization for Research and Treat- OPSCC is excellent, approximately 10%-15% will develop
ment of Cancer (EORTC) randomized studies and the distant metastases.6,8 In fact, most patients who die of HPV-
subsequent exploratory combined analysis, patients with associated OPSCC die from distant recurrence. This clinical
positive margins and extracapsular extension derive the great- observation has led to the false conclusion that patients with
est benefit from the concurrent addition of cisplatin to post- HPV-associated OPSCC have a higher rate of distant recur-
operative RT.25-27 Interestingly, chemotherapy is used less rence and that their distant recurrences are more aggressive.
often than expected after transoral surgery. This is in part These conclusions are used to support the argument that the
explained by the low rates of positive margins (see above neoadjuvant chemotherapy deintensification approach has the
section on lowering the RT dose) and absence of the “close advantage of reducing the radiation dose while still intensively
margin” historical convention. However, the incidence of treating distant metastases. However, the objective data have
nodal extracapsular extension has not changed from historical shown similar distant recurrent rates for HPV-negative and
en bloc open surgeries. For example, only 16% of patients in HPV-positive OPSCC6,8,29 and that the survival after develop-
the multicenter TLM study received adjuvant CRT, even ment of distant metastases is better in patients with HPV-
though 64% had ECS and 7% had positive margins.16 Despite associated OPSCC.29
the deintensification of chemotherapy, the overall local- ECOG recently published its results of a phase II trial
regional control rate was 93%. Proponents of transoral surgery (E1308) of neoadjuvant chemotherapy followed by reduced-
do not consider ECS to be a poor prognostic factor and have dose RT and weekly cetuximab.30 Eighty patients were
published retrospective analyses showing that the presence of enrolled. Patients received 3 cycles of neoadjuvant
30 B.S. Chera and R.J. Amdur

chemotherapy (cisplatin 75 mg/m2, paclitaxel 90 mg/m2, and The efficacy of cetuximab in HPV-associated OPSCC is
cetuximab 250 mg/m2) followed by clinical response adjusted also being questioned. Epidermal growth factor receptor
RT with concurrent cetuximab. If patients had a complete expression is lower in HPV-positive OPSCC as compared
clinical response to neoadjuvant chemotherapy at the primary to HPV-negative OPSCC,33,34 and emerging data suggest
site (manual palpation and endoscopic evaluation) or nodal that cetuximab may be less efficacious than cytotoxic
sites (palpation), they received reduced-dose RT to 54 Gy. If chemotherapy when combined with definitive RT.32,35,36
there was a partial clinical response, the RT dose was 69.3 Gy.
RT dose specification was done separately for primary and
nodal sites: for example, patient could have complete response Proton Paradigm
at the primary site which would receive 54 Gy and a partial
response in the neck which would receive 69.3 Gy. Seventy The promise of protons is in its dosimetric advantages of
percentage of patients had complete responses at the primary reducing the low-radiation dose beam path-related toxicity,
site and 58% at the nodal sites. Thirteen (16%) patients namely reductions in anterior oral cavity and posterior neck/
received the incorrect RT doses (protocol violations): for brainstem/brain incidental irradiation.37,38 It is reasonable to
example, patient with complete response at primary site hypothesize that the dosimetric benefits of protons could
received 69.3 Gy. With a median follow-up of 35 months, translate to lower symptom burden and better quality of life in
the 2-year progression-free survival (PFS) and OS was 78% patients with HPV-associated OPSCC. Early, preliminary data
and 91%, respectively, for the intent to treat analysis. All from a retrospective comparison of patients treated at MD
recurrences occurred in patients with 410 pack years smok- Anderson Cancer Center with IMRT vs intensity-modulated
ing history. The subset of patients with the best outcomes were proton therapy showed no clinically meaningful differences in
those with ≤10 pack years and oT4, N2c (2-year PFS 95%, patient-reported outcomes. Unfortunately, mucositis, pain,
2-year OS 95%). xerostomia, and dysphagia, the most important and ubiqui-
The authors do not provide details on how many patients tous toxicities oropharyngeal cancer patients experience, were
were able to receive reduced-dose RT to both the primary and similar between the 2 cohorts.37,38 We interpret these negative
nodal sites. It is likely that there were very few patients who had results to support the rationalization that it is the total-dose and
a complete response in both their primary and nodal sites and high-dose volume that are most associated with toxicity, and
thus truly received deintensified RT. Complete response rates that deintensification efforts that reduce the total-dose and
to neoadjuvant chemotherapy did not correlate with smoking high-dose treatment volumes will have the biggest impact. MD
status (complete response rate was only 45% in never Anderson Cancer Center and others are currently conducting a
smokers). This study does not answer the question whether randomized control trial to further assess the possible benefits
neoadjuvant chemotherapy is needed to deintensify RT—is this of proton therapy in patients with OPSCC (NCT01893307
trade-off necessary? clinicaltrials.gov).

Reduction Elective Nodal


Targeted Therapy Paradigm Radiation Volume or Dose
Cetuximab, in conjunction with RT, has been shown to
Paradigm
improve survival in locally advanced head and neck cancer Omitting contralateral neck radiation significantly improves
while not increasing the rate of ≥grade 3 mucositis or patient-reported quality of life. Ipsilateral radiation can be
dysphagia, late effects, or worsening quality of life.31 The safely done in patients with well-lateralized tonsil primaries
prevailing opinion is that cetuximab is less toxic than cisplatin that neither invade the base of tongue nor have significant soft
and cetuximab is used, standardly, as an alternative to cisplatin. palate invasion (extent of invasion is ≥1 cm away from the
As a way to deintensify therapy, NRG Oncology uvula).39 Though the extent of the primary tonsil cancer
(NCT01302834) and Tasman Radiation Oncology Group dictates the risk of contralateral nodal involvement, most of the
(TROG) (NCT01855451) are conducting phase III studies patients reported in the retrospective unilateral tonsil irradi-
evaluating the noninferiority of cetuximab compared with ation studies had N0 to N2b disease, with the majority having
high-dose cisplatin in HPV-associated OPSCC. The RT dose N0 to N2a disease and those with N2b having low volume of
will not be decreased in this study; thus, the major long-term neck disease.39-42 These data were generated in the pre-HPV
toxicities related to RT (ie, xerostomia and dysphagia) may not era; they are applicable to patients with well-lateralized HPV-
be reduced. associated OPSCC. Though the burden of nodal disease is
The assertion that concurrent cetuximab is less toxic than higher in HPV-associated OPSCC, the increase in N-stage
concurrent chemotherapy is being challenged. An Italian designation has primarily been N2a-N2b (not bilateral, N2c) in
randomized phase II study of RT with concurrent weekly the HPV era, suggesting that regional spread patterns have not
cisplatin 40 mg/m2 vs weekly cetuximab 250 mg/m2 showed changed and, in appropriately selected patients, ipsilateral RT
similar rates of ≥grade 3 mucositis between the 2 arms, with is safe.43
more treatment breaks (410 days) and severe or fatal adverse Nevens et al44 have conducted a randomized study inves-
events associated with the cetuximab arm.32 tigating the reduction of the dose to elective nodal levels. Two
Treatment deintensification in HPV-associated OPSCC carcinoma 31

hundred patients were randomized to either 50 Gy (control) or between good and nonresponders. Long-term patient-
40 Gy (experimental) elective nodal radiation doses. Both reported quality of life and xerostomia scores were not
nonoropharynx and oropharynx primaries were allowed, clinically significantly different.
and only ~10% of enrolled patients had HPV-associated
OPSCC. The primary endpoint was dysphagia at 6 months,
which was not significantly (clinically or statistically) different Radiation-Alone Paradigm
between the 2 arms. The local failure rate was similar between
each arm; however, the regional failure rate was 5.5% in the There are multiple studies demonstrating very high cure rates
50 Gy arm vs 13.4% in the 40 Gy arm (P ¼ 0.08). with RT alone in patients with stage T1-2, N0-1 OPSCC with
These same researchers have also presented preliminary data and without consideration of HPV or smoking status.8,47
on another randomized phase II study investigating a reduc- Because of the enhanced radiocurability of HPV-associated
tion in the irradiated volume of the elective neck.45 Based on OPSCC, omitting chemotherapy even for higher-staged
the results of the aforementioned study, the elective nodal patients is another approach that has been proposed to
radiation dose was 40 Gy. In the experimental arm, contrala- deintensify therapy.48 This may be more appropriate for the
teral levels 3 and 4 were omitted. One hundred patients were more favorable risk patients, specifically those with ≤ 10 pack
randomized, and both oropharynx and nonoropharynx pri- years smoking histories.48
mary sites were allowed. Fifty percentage of enrolled patients The concern of a radiation-alone approach is that the occult
had OPSCC; however, the HPV status was unknown for most distant metastatic disease is not being treated. Though it is
of them. Primary objective was toxicity at 6 and 12 months, debatable whether systemic chemotherapy is effective at
which were not significantly different (grade ≥ 2 dysphagia, treating micrometastatic distant disease, in the HPV-associated
xerostomia, and fibrosis). With a minimum follow-up of OPSCC subset, it may be more efficacious as the response rate
1 year, there were no statistical difference in local control to chemotherapy is better.
(86% vs 90%, P 4 0.05) or regional control (84% vs 92%,
P 4 0.05), with 8 regional failures (1 in the nontreated elective
neck) in the experimental arm vs 4 in the control arm. University of North Carolina/
Taken together, these 2 prospective studies provide insuffi-
cient data to support the use of reducing the elective nodal
University of Florida Paradigm
radiation dose and volume as a means of deintensification. Our approach has been to substantially reduce both RT and
Long-term toxicity was not significantly reduced, and though chemotherapy without using definitive surgery or induction
not statistically significant, there were more regional failures in chemotherapy. In our first phase II study, we enrolled 44
the electively irradiated neck. patients with HPV-associated OPSCC, of which 64% were
The University of Chicago has investigated the use of HPV and p16 positive and 82% were never smokers.49 Patients
intensive induction chemotherapy followed by limiting radi- with T4 and N3 disease were not eligible. All patients received
ation volume to the original sites of gross disease at the primary 60 Gy (2 Gy/fx, daily) IMRT with concurrent weekly cisplatin
site and neck.46 Ninety-four patients were enrolled in a phase of 30 mg/m2. Compared to the standard of 70 Gy and
2 clinical trial and 59 patients (76%) had OPSCC of which cumulative cisplatin dose of 300 mg/m2, radiation was
83% were HPV-associated. All patients received induction reduced to 10 Gy and cisplatin by 40%. The primary endpoint
chemotherapy (cisplatin, paclitaxel, cetuximab, and was pathological complete response (pCR), and all patients
±evorlimus) followed by THFX (paclitaxel, fluorouracil, had a biopsy of their primary site and supraselective neck
hydroxyurea, and 1.5 Gy twice daily RT every other week). dissections (nodal pluck or removal of initial involved nodal
Patients who were good responders to induction chemo- level) 6-14 weeks after completing deintensified CRT. Our null
therapy had 450% radiographic response to targeted lesions. hypothesis was that the pCR rate would be ~87% (the reported
Patients with good response to chemotherapy only had their 3-year local-regional control rate in RTOG 0129).6 We
gross tumor (plus 1.5 cm margin) treated to 75 Gy (no elective observed a pCR rate of 86% (98% at the primary site and
nodal radiation). Patients with ≤50% (nonresponders) radio- 84% in the neck).49 With a median follow-up of 36 months
graphic response had their gross tumor treated to 75 Gy and (88% have at least 2 years follow-up), the 2-year OS is 95%
the first uninvolved nodal echelon to a dose of 45 Gy. Thirty- (1 patient died from cerebrovascular accident and another died
seven of 89 assessable patients had good responses to from a glioblastoma multiforme) and 2-year cause-specific
induction chemotherapy, 30 of whom had HPV-associated survival is 100%. Six patients had partial pathological
OPSCC (ie, 30/59, 51% of HPV-associated OPSCC had good response, all of which were micrometastatic (o1 mm) resid-
response). With a mean follow-up of 2 years, the 2-year PFS ual, equivocal foci. One of these 6 patients had an equivocal
and OS for HPV-associated OPSCC with good response were focus in the base of tongue (biopsy) and neck node for which
93.1% and 92.1%, respectively. Two-year PFS and OS for he had a transoral resection (no further additional neck
HPV-associated OPSCC with ≤ good response were 74.0% dissection) showing no residual cancer. The other 5 patients
and 95.2%, respectively. As far as toxicity, patients with good only had microscopic residual in a lymph node(s) and did not
response were less likely to have percutaneous endoscopic have further surgery. All 6 patients are alive with no evidence of
gastrostomy placement during treatment, though long-term disease. Global quality of life (assessed by EORTC QLQ C30)
percutaneous endoscopic gastrostomy dependence was similar returned to baseline and patients reported (assessed by
32 B.S. Chera and R.J. Amdur

PRO-CTCAE) mild-to-moderate xerostomia and none-to-mild potential adjunctive biomarkers that can help us better select
dysphagia at 24 months posttreatment. Thirty-nine percentage patients for deintensification.
required a feeding tube (median duration of 15 weeks) Another area of future research would be adaptive treatment
of which none were permanent. The results of our first study using imaging biological correlates early during treatment to
are encouraging as the pCR was very high and the 2-year help in deciding when to deintensify. Lee and colleagues
clinical outcomes and patient-reported outcome were excel- performed a pilot study in 33 patients in whom they
lent. Based on our results, the NRG oncology group is performed pretreatment 18F-fluorodeoxyglucose and
conducting a phase II randomized trial comparing 60 Gy dynamic18F-fluoromisonidazole positron emission tomo-
IMRT with concurrent weekly cisplatin 40 mg/m2 (6 weeks) graphy (PET) scans. For patients with pretreatment hypoxia
vs accelerated 60 Gy IMRT (5 weeks) (NCT02254278, on 18F-fluoromisonidazole PET, a repeat scan was done
clinicaltrials.gov). 1 week after chemoradiation. Patients without pretreatment
We have completed accrual (n ¼ 100) to our second- hypoxia or with resolution of hypoxia on repeat scan
generation phase II trial that has the more traditional endpoint received a 10-Gy dose reduction to metastatic lymph node
of 2-year PFS. A 12-week post deintensified CRT PET/CT is (s). All 33 patients had pretreatment hypoxia, and 30% met
used to guide surgical evaluation. Other major differences criteria for 10 Gy dose reduction to lymph node(s). With a
included the following: (1) chemotherapy is omitted in median follow-up of 32 months, the 2-year local-regional
patients with T0-T2, N0-N1 stages (ie, 60 Gy IMRT alone), control rate was 100%.
(2) patients with moderate, remote smoking histories are Early results have shown that deintensification is possible in
eligible (≤30 pack years and 5 years abstinence), and carefully selected patients; however, the optimal level of
(3) genomic sequencing of pretreatment biopsies are deintensification is not known. Can we go lower than 60 Gy
performed. in the definitive setting (without induction) or lower than
Our ongoing third-generation deintensification phase II 54 Gy after induction chemotherapy? Assessment of bio-
study is investigating whether tumor genomics can be used markers pretreatment (eg, tumor genomics) and during treat-
to select patients with 410 pack years histories in whom ment (eg, circulating tumor DNA, tumor hypoxia using
deintensification may be safely done. Patients with HPV- functional radiographic imaging techniques) may allow us to
associated OPSCC regardless of smoking history are eligible super-select patients in whom it may be safe to undergo ultra-
(excluding T4 and N3). Tumor genomics are performed on deintensification.
tumor samples of patients who have 410 pack years smoking Future studies may not be able to define the optimal
histories and if P53 is wild-type, they will receive deintensified deintensification paradigm or strategy. Patient, physician,
CRT (60 Gy IMRT þ weekly cisplatin 30 mg/m2). The results and institutional biases will influence the field of deintensified
of the genetic analysis is available by week 2 or 3 of radiation, treatments for HPV-associated OPSCC, and just as there are
and if P53 is found to be mutated, patients will receive an now 3 major “preferred” treatment paradigms for oropharynx
additional 10 Gy and another week of cisplatin 30 mg/m2 cancer (surgery first, radiation first, and induction first), in the
(70 Gy IMRT þ cumulative cisplatin 200 mg/m2). Mutations future (we project) there will be 3 acceptable deintensification
in P53 are rarely seen in HPV-associated OPSCC and are paradigms. Purist will call for phase III randomized controlled
indicative that tobacco may have been the major driver of trials to find the best paradigm. Conducting these gold
carcinogenesis.50 We are also prospectively analyzing plasma standard studies may be difficult as patient aversion to
circulating HPV tumor DNA (baseline, weekly during treat- randomization and their desire for faster discovery of newer
ment, and with each follow-up visit) to evaluate its predictive treatments increase and physicians growing lack of equipoise
and prognostic value. influence their willingness to participate in phase III studies.

Future Directions Conclusions


There are several questions that remain to be answered. We There are several deintensified treatment strategies for HPV-
now have a biomarker that we can reliably use in the practice of associated OPSCC, each with their unique caveats and
head and neck oncology, but it is not perfect. Though HPV controversies. There are several phase II studies with excellent
status, smoking status, and T and N stage do a “pretty good” outcomes. In the near future, there will be several accepted
job at providing prognostic information and define risk groups deintensified approaches centered around (1) transoral surgi-
in a way that we can (with relative confidence) select patients cal followed by pathological risk-adapted, reduced-dose radi-
who would most benefit (ie, not compromise tumor control) ation and chemotherapy, (2) neoadjuvant chemotherapy
from deintensification—there are still patients with favorable followed by reduced-dose (and possibly volume) radiation,
risk disease (≤10 pack years) who do not experience cancer and (3) reduced-dose radiation ± chemotherapy. Bastions of
control and there are patients with unfavorable risk disease excellence in surgery, chemotherapy, and radiation will favor
(≥ 10 pack years, T4, N3) who may benefit from deintensi- one of these 3 paradigms. Though the results of large
fication. Our ability to risk stratify can be refined with the use randomized studies are often required to change medical
of other biomarkers. In our third-generation phase II, we are practice, patient expectations as health care consumers (risk
studying tumor genomics and circulating tumor DNA as adverse to randomization) and physician biases (lack of
Treatment deintensification in HPV-associated OPSCC carcinoma 33

equipoise) may impede the conduction of traditional phase III 20. Bussels B, Hermans R, Reijnders A, et al: Retropharyngeal nodes in
squamous cell carcinoma of oropharynx: Incidence, localization, and
trials. Further, the excellent results of well conducted phase II
implications for target volume. Int J Radiat Oncol Biol Phys 65
studies may be enough to change medical practice. (3):733-738, 2006
21. Hinni ML, Zarka MA, Hoxworth JM: Margin mapping in transoral surgery
References for head and neck cancer. Laryngoscope 123(5):1190-1198, 2013
22. Fried D, Lehman-Davis M, Willson A, et al: Dosimetric feasibility of
1. Chaturvedi AK, Engels EA, Pfeiffer RM, et al: Human papillomavirus and
sparing the primary site for oropharyngeal squamous cell carcinoma after
rising oropharyngeal cancer incidence in the United States. J Clin Oncol
transoral laser microsurgery in patients with unilateral positive neck
29(32):4294-4301, 2011
nodes. Pract Radiat Oncol 3(4):282-286, 2013
2. D’Souza G, Kreimer AR, Viscidi R, et al: Case-control study of human
23. Eisbruch A, Kim HM, Feng FY, et al: Chemo-IMRT of oropharyngeal
papillomavirus and oropharyngeal cancer. N Engl J Med 356
cancer aiming to reduce dysphagia: Swallowing organs late complication
(19):1944-1956, 2007
probabilities and dosimetric correlates. Int J Radiat Oncol Biol Phys 81(3):
3. Mork J, Lie AK, Glattre E, et al: Human papillomavirus infection as a risk
e93-e99, 2011
factor for squamous-cell carcinoma of the head and neck. N Engl J Med
24. Feng FY, Kim HM, Lyden TH, et al: Intensity-modulated chemoradio-
344(15):1125-1131, 2001
therapy aiming to reduce dysphagia in patients with oropharyngeal
4. Fakhry C, Westra WH, Li S, et al: Improved survival of patients with
cancer: Clinical and functional results. J Clin Oncol 28(16):2732-2738,
human papillomavirus-positive head and neck squamous cell carcinoma
2010
in a prospective clinical trial. J Natl Cancer Inst 100(4):261-269, 2008
25. Bernier J, Cooper JS, Pajak TF, et al: Defining risk levels in locally
5. Lassen P, Eriksen JG, Hamilton-Dutoit S, et al: Effect of HPV-associated
advanced head and neck cancers: A comparative analysis of concurrent
p16INK4A expression on response to radiotherapy and survival in
postoperative radiation plus chemotherapy trials of the EORTC (#22931)
squamous cell carcinoma of the head and neck. J Clin Oncol 27
and RTOG (# 9501). Head Neck 27(10):843-850, 2005
(12):1992-1998, 2009
26. Bernier J, Domenge C, Ozsahin M, et al: Postoperative irradiation with or
6. Ang KK, Harris J, Wheeler R, et al: Human papillomavirus and survival of
without concomitant chemotherapy for locally advanced head and neck
patients with oropharyngeal cancer. N Engl J Med 363(1):24-35, 2010
cancer. N Engl J Med 350(19):1945-1952, 2004
7. Licitra L, Perrone F, Bossi P, et al: High-risk human papillomavirus affects
prognosis in patients with surgically treated oropharyngeal squamous cell 27. Cooper JS, Pajak TF, Forastiere AA, et al: Postoperative concurrent
carcinoma. J Clin Oncol 24(36):5630-5636, 2006 radiotherapy and chemotherapy for high-risk squamous-cell carcinoma of
8. O’Sullivan B, Huang SH, Siu LL, et al: Deintensification candidate the head and neck. N Engl J Med 350(19):1937-1944, 2004
subgroups in human papillomavirus-related oropharyngeal cancer 28. Sinha P, Lewis Jr. JS, Piccirillo JF, et al: Extracapsular spread and adjuvant
according to minimal risk of distant metastasis. J Clin Oncol 31 therapy in human papillomavirus-related, p16-positive oropharyngeal
(5):543-550, 2013 carcinoma. Cancer 118(14):3519-3530, 2012
9. Haddad R, O’Neill A, Rabinowits G, et al: Induction chemotherapy 29. Huang SH, Perez-Ordonez B, Weinreb I, et al: Natural course of distant
followed by concurrent chemoradiotherapy (sequential chemoradiother- metastases following radiotherapy or chemoradiotherapy in HPV-related
apy) versus concurrent chemoradiotherapy alone in locally advanced oropharyngeal cancer. Oral Oncol 49(1):79-85, 2013
head and neck cancer (PARADIGM): A randomised phase 3 trial. Lancet 30. Marur S, Li S, Cmelak AJ, et al: E1308: Phase II trial of induction
Oncol 14(3):257-264, 2013 chemotherapy followed by reduced-dose radiation and weekly cetuximab
10. Posner MR, Hershock DM, Blajman CR, et al: Cisplatin and fluorouracil in patients with HPV-associated resectable squamous cell carcinoma of the
alone or with docetaxel in head and neck cancer. N Engl J Med 357 oropharynx—ECOG-ACRIN Cancer Research Group. J Clin Oncol 2016.
(17):1705-1715, 2007 JCO2016683300
11. Cohen EE, Karrison TG, Kocherginsky M, et al: Phase III randomized trial 31. Bonner JA, Harari PM, Giralt J, et al: Radiotherapy plus cetuximab for
of induction chemotherapy in patients with N2 or N3 locally advanced locoregionally advanced head and neck cancer: 5-year survival data from a
head and neck cancer. J Clin Oncol 32(25):2735-2743, 2014 phase 3 randomised trial, and relation between cetuximab-induced rash
12. Machtay M, Moughan J, Trotti A, et al: Factors associated with severe late and survival. Lancet Oncol 11(1):21-28, 2010
toxicity after concurrent chemoradiation for locally advanced head and 32. Magrini SM, Buglione M, Corvo R, et al: Cetuximab and radiotherapy
neck cancer: An RTOG analysis. J Clin Oncol 26(21):3582-3589, 2008 versus cisplatin and radiotherapy for locally advanced head and neck
13. Parsons JT, Mendenhall WM, Stringer SP, et al: Squamous cell carcinoma cancer: A randomized phase II trial. J Clin Oncol 34(5):427-435, 2016
of the oropharynx: Surgery, radiation therapy, or both. Cancer 94 33. Hong A, Dobbins T, Lee CS, et al: Relationships between epidermal
(11):2967-2980, 2002 growth factor receptor expression and human papillomavirus status as
14. Patel SC, Hackman T, Hayes DN, et al: De-intensification of treatment for markers of prognosis in oropharyngeal cancer. Eur J Cancer 46
human papilloma virus associated oropharyngeal squamous cell carci- (11):2088-2096, 2010
noma: A discussion of current approaches. Pract Radiat Oncol 2 34. Kong CS, Narasimhan B, Cao H, et al: The relationship between human
(4):282-287, 2012 papillomavirus status and other molecular prognostic markers in head
15. Weinstein GS, Quon H, O’Malley Jr. BW, et al: Selective neck dissection and neck squamous cell carcinomas. Int J Radiat Oncol Biol Phys 74
and deintensified postoperative radiation and chemotherapy for orophar- (2):553-561, 2009
yngeal cancer: A subset analysis of the University of Pennsylvania transoral 35. Ley J, Mehan P, Wildes TM, et al: Cisplatin versus cetuximab given
robotic surgery trial. Laryngoscope 120(9):1749-1755, 2010 concurrently with definitive radiation therapy for locally advanced head
16. Haughey BH, Hinni ML, Salassa JR, et al: Transoral laser microsurgery as and neck squamous cell carcinoma. Oncology 85(5):290-296, 2013
primary treatment for advanced-stage oropharyngeal cancer: A United 36. Koutcher L, Sherman E, Fury M, et al: Concurrent cisplatin and radiation
States multicenter study. Head Neck 33(12):1683-1694, 2011 versus cetuximab and radiation for locally advanced head-and-neck
17. Rich JT, Milov S, Lewis Jr. JS, et al: Transoral laser microsurgery (TLM) þ/- cancer. Int J Radiat Oncol Biol Phys 81(4):915-922, 2011
adjuvant therapy for advanced stage oropharyngeal cancer: Outcomes and 37. Sio TT, Lin HK, Shi Q, et al: Intensity modulated proton therapy versus
prognostic factors. Laryngoscope 119(9):1709-1719, 2009 intensity modulated photon radiation therapy for oropharyngeal cancer:
18. Holsinger FC, Ferris RL: Transoral endoscopic head and neck surgery First comparative results of patient-reported outcomes. Int J Radiat Oncol
and its role within the multidisciplinary treatment paradigm of Biol Phys 95(4):1107-1114, 2016
oropharynx cancer: Robotics, lasers, and clinical trials. J Clin Oncol 38. Beitler JJ, Chera BS: Protons for oropharyngeal cancer have not yet justified
33(29):3285-3292, 2015 their promise. Int J Radiat Oncol Biol Phys 95(4):1115-1116, 2016
19. Grant DG, Hinni ML, Salassa JR, et al: Oropharyngeal cancer: A case for 39. O’Sullivan B, Warde P, Grice B, et al: The benefits and pitfalls of ipsilateral
single modality treatment with transoral laser microsurgery. Arch radiotherapy in carcinoma of the tonsillar region. Int J Radiat Oncol Biol
Otolaryngol Head Neck Surg 135(12):1225-1230, 2009 Phys 51(2):332-343, 2001
34 B.S. Chera and R.J. Amdur

40. Jackson SM, Hay JH, Flores AD, et al: Cancer of the tonsil: The 45. Schatteman J, Nevens D, Nuyts S, et al: OC-0452: Prospective random-
results of ipsilateral radiation treatment. Radiother Oncol 51 ized adaptive dose-de-escalation in the elective neck: Late toxicity and
(2):123-128, 1999 control. Radiother Oncol 119:S211-S212, 2016
41. Kagei K, Shirato H, Nishioka T, et al: Ipsilateral irradiation for carcinomas 46. Villaflor VM, Melotek JM, Karrison TG, et al: Response-adapted volume
of tonsillar region and soft palate based on computed tomographic de-escalation (RAVD) in locally advanced head and neck cancer. Ann
simulation. Radiother Oncol 54(2):117-121, 2000 Oncol 27(5):908-913, 2016
42. Rusthoven KE, Raben D, Schneider C, et al: Freedom from local and 47. Garden AS, Asper JA, Morrison WH, et al: Is concurrent chemoradiation
the treatment of choice for all patients with Stage III or IV head and neck
regional failure of contralateral neck with ipsilateral neck radio-
carcinoma? Cancer 100(6):1171-1178, 2004
therapy for node-positive tonsil cancer: Results of a prospective
48. O’Sullivan B, Huang SH, Perez-Ordonez B, et al: Outcomes of HPV-
management approach. Int J Radiat Oncol Biol Phys 74(5):
related oropharyngeal cancer patients treated by radiotherapy alone using
1365-1370, 2009
altered fractionation. Radiother Oncol 103(1):49-56, 2012
43. Galloway TJ, Lango MN, Burtness B, et al: Unilateral neck therapy in the 49. Chera BS, Amdur RJ, Tepper J, et al: Phase 2 trial of de-intensified
human papillomavirus ERA: Accepted regional spread patterns. Head chemoradiation therapy for favorable-risk human papillomavirus-associ-
Neck 35(2):160-164, 2013 ated oropharyngeal squamous cell carcinoma. Int J Radiat Oncol Biol Phys
44. Nevens D, Duprez F, Daisne JF, et al: Reduction of the dose of 93(5):976-985, 2015
radiotherapy to the elective neck in head and neck squamous cell 50. Cancer Genome Atlas Network. Comprehensive genomic characterization
carcinoma; a randomized clinical trial. Effect on late toxicity and tumor of head and neck squamous cell carcinomas. Nature 517(7536):576-582,
control. Radiother Oncol 122(2):171-177, 2017 2015

You might also like