You are on page 1of 19

CAP Laboratory Improvement Programs

The Cancer Immunotherapy Biomarker Testing Landscape


Eric E. Walk, MD; Sophia L. Yohe, MD; Amy Beckman, MD; Andrew Schade, MD, PhD; Mary M. Zutter, MD;
John Pfeifer, MD, PhD; Anna B. Berry, MD; on behalf of the College of American Pathologists Personalized Health Care Committee

 Context.—Cancer immunotherapy provides unprece- clinical trial data representing the current field of cancer
dented rates of durable clinical benefit to late-stage cancer immunotherapy.
patients across many tumor types, but there remains a Conclusions.—The cancer immunotherapy field, includ-
critical need for biomarkers to accurately predict clinical ing the use of biomarker testing to predict patient
response. Although some cancer immunotherapy tests are response, is still in evolution. PD-L1, mismatch repair,
associated with approved therapies and considered vali- and microsatellite instability testing are helping to guide
dated, other biomarkers are still emerging and at various the use of US Food and Drug Administration–approved
states of clinical and translational exploration. therapies, but there remains a need for better predictors of
Objective.—To provide pathologists with a current and response and resistance. Several categories of tumor and
practical update on the evolving field of cancer immuno-
patient characteristics underlying immune responsiveness
therapy testing. The scientific background, clinical data,
and testing methodology for the following cancer immu- are emerging and may represent the next generation of
notherapy biomarkers are reviewed: programmed death cancer immunotherapy predictive biomarkers. Pathologists
ligand-1 (PD-L1), mismatch repair, microsatellite instabil- have important roles and responsibilities as the field of
ity, tumor mutational burden, polymerase d and e cancer immunotherapy continues to develop, including
mutations, cancer neoantigens, tumor-infiltrating lympho- leadership of translational studies, exploration of novel
cytes, transcriptional signatures of immune responsiveness, biomarkers, and the accurate and timely implementation
cancer immunotherapy resistance biomarkers, and the of newly approved and validated companion diagnostics.
microbiome. (Arch Pathol Lab Med. 2020;144:706–724; doi: 10.5858/
Data Sources.—Selected scientific publications and arpa.2018-0584-CP)

C ancer immunotherapy has revolutionized the field of


oncology by delivering unprecedented levels of durable
mechanisms, can efficiently eradicate even widespread
metastatic cancer.1,2 This realization has had a transforma-
tive impact on the fields of oncology, radiology, and cancer
survival benefit for cancer patients, including some patients
drug development, as evidenced by new first-line treatment
with previously incurable late-stage disease. It is now widely
options, new criteria for radiologic response,3 and dramatic
accepted that the human immune system, when properly shifts in pharmaceutical pipeline strategies. The US Food
activated and in the absence of negative regulatory and Drug Administration (FDA) has approved multiple
cancer immunotherapies for a wide range of cancer
indications (Table 1),4 and more than 2000 cancer immu-
Accepted for publication August 23, 2019.
Published online November 12, 2019. notherapy agents are currently in clinical development.5
From the Department of Medical & Scientific Affairs, Roche Tissue Despite all the enthusiasm and positive clinical outcome
Diagnostics, Tucson, Arizona (Dr Walk); the Department of data, however, cancer immunotherapy currently benefits
Laboratory Medicine and Pathology, University of Minnesota only a small subset of cancer patients—around 20% on
Medical School, Minneapolis (Drs Yohe and Beckman); Diagnostic average across the cancer indications assessed through
and Experimental Pathology, Eli Lilly and Company, Indianapolis,
Indiana (Dr Schade); the Department of Pathology, Microbiology,
clinical trials.6 Because not all patients respond to cancer
and Immunology, Vanderbilt University School of Medicine, Nash- immunotherapy, and some experience serious adverse
ville, Tennessee (Dr Zutter); the Department of Molecular Pathology immune reactions,7 biomarkers predicting efficacy are
and Genomics, Swedish Cancer Institute, Seattle, Washington (Dr critically needed both for current clinical care and to enable
Berry); and the Department of Pathology, Washington University and drive further progress in this rapidly advancing field.
School of Medicine, St Louis, Missouri (Dr Pfeifer).
Drs Pfeifer and Berry are co–senior authors.
Anatomic and molecular pathologists have the opportu-
Dr Walk is an employee at Roche Tissue Diagnostics and has stock nity to be at the center of the development, validation, and
ownership. Dr Schade is an employee at Eli Lilly and Company. The clinical implementation of patient selection biomarkers for
other authors have no relevant financial interest in the products or cancer immunotherapy. Predictive immunotherapy bio-
companies described in this article. markers such as programmed death ligand-1 (PD-L1)
All authors are past or current members of the College of American immunohistochemistry (IHC), mismatch repair (MMR)
Pathologists Personalized Health Care Committee.
Corresponding author: Eric E. Walk, MD, Department of Medical IHC, and microsatellite instability (MSI) testing are already
& Scientific Affairs, Roche Tissue Diagnostics, 1910 E Innovation established as routine in many pathology laboratories
Park Dr, Tucson, AZ 85718 (email: eric.walk@roche.com). around the world. In addition, emerging cancer immuno-
706 Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al
therapy biomarkers such as tumor-infiltrating lymphocyte cells then traffic back to the tumor to find and destroy their
(TIL) assessment and multiplexed assessment of the tumor cancer cell targets, triggering release of additional tumor
microenvironment are dependent on in situ cellular and neoantigens and renewing the cancer immunity cycle.
histopathologic interpretation. Taken together, it is clear Interruption of the cancer immunity cycle can clearly
that optimal progress in the field of cancer immunotherapy occur and results in immune evasion, uncontrolled tumor
would benefit from, and, one could argue, is dependent growth, and clinical progression. The success of cancer
upon, pathologist leadership and stewardship. immunotherapy to date has come from a careful elucidation
Clear guidelines for cancer immunotherapy testing are not of this cycle, including the specific parameters and
currently available, and the field is evolving rapidly in mechanisms underlying immune responsiveness and resis-
response to new clinical and translational data. The intent of tance, an approach that will continue to be necessary for
this article is to briefly review the current landscape of further advancement of the field. The first of the cancer
cancer immunotherapy biomarker testing as a resource for immune mechanisms to be understood and successfully
individualized test selection in clinical pathology practice targeted were the immune regulatory checkpoints.
and to provide an overview of this complex, ever-changing A complex system of regulatory checkpoints has evolved in
field. Included are those cancer immunotherapy tests humans to ensure immune homeostasis and prevent
currently accepted as standard of care for clinical testing, autoimmunity. Central to this system of regulation are
including those assays with FDA approval as well as immune checkpoint proteins present on T lymphocytes and
emerging assays and technologies still undergoing investi- antigen-presenting cells. More than a dozen immune
gation and clinical validation. For each biomarker, the checkpoint proteins have been discovered, some functioning
clinical adoption status, scientific background, available to activate the immune system and others functioning to
clinical data, and testing methodology will be reviewed. down-regulate it.13 The pioneering work of Jim Allison, PhD
This article is focused on providing a general awareness of (MD Anderson Cancer Center, Houston, Texas), led to the
the current testing landscape. It is not meant to be a discovery that cytotoxic T-lymphocyte associated protein 4
complete review of the entire cancer immunotherapy field, (CTLA-4) acts as an inhibitory checkpoint molecule, func-
nor is it meant to serve as an official guideline for tioning as a brake on antitumor immune responses.14 Dr
immunotherapy testing or to provide clinical treatment Allison hypothesized that inhibiting CTLA-4 via antibody
recommendations. blockade would abrogate suppression of immune priming
and enhance the immune response to cancer.14 Initial positive
BACKGROUND AND HISTORY OF CANCER results in animals led to subsequent clinical validation of
IMMUNOTHERAPY CTLA-4 inhibition in metastatic melanoma and to eventual
FDA approval of ipilimumab in 2011.15 Dr Allison and Tasuku
The immune system is divided into 2 distinct response
Honjo, MD, PhD (Kyoto University, Kyoto, Japan) were
components, innate and adaptive. The innate immune
jointly awarded the 2018 Nobel Prize in Physiology or
system responds rapidly but indiscriminately and includes
Medicine for their discovery of cancer therapy by inhibition of
neutrophils and macrophages, whose receptors recognize
negative immune regulation.16 In 2014, the FDA approved
foreign antigens such as microbial products. In contrast, the
nivolumab, which inhibits the programmed death receptor-1
adaptive immune response is highly targeted and precise,
(PD-1), a checkpoint molecule that directly regulates T-cell
with a vast antigen response diversity achieved through the
effector activity in the tumor microenvironment. There are
rearrangement of B-lymphocyte immunoglobulin genes and
currently several FDA-approved checkpoint inhibitor thera-
T-lymphocyte antigen receptor genes.8 Although slower to pies targeting CTLA-4 as well as PD-1 and its ligand, PD-L1
develop, the incredible selectivity of the adaptive immune (Table 1),17,18 with numerous additional immune checkpoint
system appears to underlie the clinical effectiveness of inhibitors in clinical development.5
cancer immunotherapy.
Although associations between infections and cancer PD-L1 (STATUS: FDA-APPROVED THERAPIES AND
regression had been made as early as the 1700s, William ASSAYS)
Coley, MD, is widely acknowledged as the ‘‘Father of
Immunotherapy’’ because of his systematic, lifelong study of PD-L1 Biology and Anti–PD-(L)1 Therapies
induced bacterial infections as therapy for sarcoma patients, The PD-L1 ligand and its cognate receptor, PD-1, are
beginning in 1891.9 Although Coley firmly believed in the components of an immune inhibitory axis that, when
tie between severe infections and cancer regressions, the activated, negatively regulates T-cell signaling, effector
mechanisms underlying the responses he observed were not function, and killing capacity.13 Tumor cells can activate this
elucidated until the mid to late 1950s with the realization pathway through expression of PD-L1 on the cell surface via
that the immune system could recognize and attack tumors 2 distinct mechanisms, a constitutive mechanism triggered by
based on specific antigens.10,11 genomic alterations (see Table 2) and an inducible mecha-
Our current mechanistic understanding of immune-medi- nism termed adaptive immune resistance.13,19 In the latter case,
ated cancer elimination is based on the widely accepted tumor surface expression of PD-L1 occurs in response to the
process referred to as the cancer immunity cycle (Figure 1).12 release of the cytokine interferon c (IFN-c) from T cells upon
This process, which is dependent on the adaptive immune tumor recognition. In this way, tumors avoid immune-
system, is initiated when necrotic and/or apoptotic tumor mediated destruction by activating a naturally occurring
cells release immunogenic neoantigen proteins into the immune regulatory pathway. Reactivation of T-cell prolifer-
tumor microenvironment. These neoantigens are recognized ation and effector function through antibody blockade of PD-
as foreign by the immune system, and are engulfed and 1/PD-L1 has been conclusively shown to yield durable
processed by dendritic cells, which migrate through lym- clinical benefit to patients across multiple tumor types.20
phatics to lymph nodes to prime and activate tumor-specific Five anti–PD-(L)1 therapies (atezolizumab, avelumab,
cytotoxic T-cell responses. Tumor-specific CD8þ cytotoxic T durvalumab, nivolumab, and pembrolizumab) have been
Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al 707
Table 1. FDA-Approved Cancer Immunotherapies
Drug Indication
Drug Target Indication Stage Line of Therapy Diagnostic
Checkpoint inhibitors
Atezolizumab in combination PD-L1 Nonsquamous NSCLC with no EGFR Metastatic First None
with bevacizumab, or ALK genomic tumor aberrations
paclitaxel, and carboplatin
Atezolizumab PD-L1 NSCLC Metastatic Second None
Urothelial carcinoma (aa) Locally advanced or metastatic First in cisplatin ineligible FDA-approved
urothelial carcinoma PD-L1 IHC
First in platinum (any) ineligible None
Second after any platinum None
Atezolizumab in combination PD-L1 Small cell lung cancer Extensive stage First None
with carboplatin and
etoposide

708 Arch Pathol Lab Med—Vol 144, June 2020


Atezolizumab in combination PD-L1 Triple-negative breast cancer (aa) Unresectable locally advanced or First FDA-approved
with paclitaxel protein-bound metastatic PD-L1 IHC
Avelumab PD-L1 Merkel cell carcinoma (aa) Metastatic First, any None
Urothelial carcinoma (aa) Locally advanced or metastatic Second
Durvalumab PD-L1 Urothelial carcinoma (aa) Locally advanced or metastatic Second None
NSCLC Unresectable stage III that has not progressed following concurrent
platinum-based chemotherapy and radiation therapy
Ipilimumab CTLA-4 Melanoma Metastatic First None
Cutaneous melanoma, completely Lymph node positive Adjuvant
resected
Ipilimumab þ nivolumab CTLA-4 Melanoma Unresectable or metastatic First None
þ PD-1 Colorectal cancer, dMMR or MSI Metastatic Second dMMR or MSI
high (aa) high
RCC, intermediate or poor risk, Advanced First None
previously untreated
Nivolumab PD-1 Classical Hodgkin lymphoma (aa) Any Second after autologous HSCT and None
brentuximab vedotin, fourth after 3
or more lines of systemic therapy
that includes autologous HSCT
Colorectal cancer, dMMR or MSI Metastatic Second dMMR or MSI
high (aa) high
Hepatocellular carcinoma (aa) Any Second, previously treated with None
sorafenib
Melanoma Unresectable or metastatic First
Melanoma, completely resected Involvement of lymph nodes or Adjuvant
metastatic
NSCLC Metastatic Second
RCC Advanced Second after prior antiangiogenic
therapy
SCCHN Recurrent or metastatic Second
Small cell lung cancer (aa) Metastatic Second
Urothelial carcinoma (aa) Locally advanced or metastatic Second

Cancer Immunotherapy Biomarker Testing Landscape—Walk et al


Table 1. Continued
Drug Indication
Drug Target Indication Stage Line of Therapy Diagnostic
Pembrolizumab PD-1 Cervical cancer (aa) Recurrent or metastatic Second PD-L1 by FDA-
approved test
Classical Hodgkin lymphoma (aa) Refractory Fourth None
Esophageal squamous cell carcinoma Recurrent locally advanced or Second PD-L1 by FDA-
metastatic approved test
Gastric or GEJ adenocarcinoma (aa) Recurrent locally advanced or Third PD-L1 by FDA-
metastatic approved test
Hepatocellular carcinoma (aa) Previously treated with sorafenib None
HNSCC (aa) Metastatic Second None
Melanoma Unresectable or metastatic First, any

Arch Pathol Lab Med—Vol 144, June 2020


Melanoma Adjuvant treatment of patients with melanoma with involvement of lymph
node(s) following complete resection
Merkel cell carcinoma (aa) Recurrent locally advanced or First
metastatic
NSCLC with no EGFR or ALK Stage III ineligible for surgical First PD-L1 by FDA-
genomic tumor aberrations resection or definitive approved test
chemoradiation or metastatic
NSCLC Metastatic Second PD-L1 by FDA-
approved test
Primary mediastinal large B-cell Refractory Third None
lymphoma (aa)
Solid tumors, dMMR or MSI high (aa) Unresectable or metastatic Second dMMR or MSI
high
Urothelial carcinoma (aa) Locally advanced or metastatic First in cisplatin ineligible FDA-approved
PD-L1 IHC
First in platinum (any) ineligible None
Second None
Pembrolizumab in combination PD-1 RCC Advanced First None
with axitinib
Pembrolizumab in combination PD-1 Nonsquamous NSCLC with no EGFR Metastatic First None
with pemetrexed and or ALK genomic tumor aberrations
platinum chemotherapy
Pembrolizumab in combination PD-1 Squamous NSCLC Metastatic First None
with carboplatin and either
paclitaxel or paclitaxel
protein bound
Chimeric antigen receptor T-cell therapies
Axicabtagene ciloleucel CD19 Large B-cell lymphoma Relapsed/refractory Third None
Tisagenlecleucel CD19 Pediatric and young adult B-cell Relapsed/refractory Relapsed/refractory None
precursor ALL
Adult diffuse large-B cell lymphoma Relapsed/refractory Third
Abbreviations: aa, indication approved under FDA-accelerated approval (continued approval may be contingent upon verification and description of clinical benefit in confirmatory trials); ALK,
anaplastic lymphoma kinase; ALL, acute lymphoblastic leukemia; CTLA-4, cytotoxic T-lymphocyte associated protein 4; dMMR, mismatch repair deficient; EGFR, epidermal growth factor receptor;
FDA, US Food & Drug Administration; GEJ, gastroesophageal junction; HNSCC, squamous cell carcinoma of the head and neck; HSCT, hematopoietic stem cell transplantation; IHC,
immunohistochemistry; MSI, microsatellite instability; NSCLC, non–small cell lung carcinoma; PD-1, programmed death receptor-1; PD-L1, programmed death ligand-1; RCC, renal cell carcinoma;

Cancer Immunotherapy Biomarker Testing Landscape—Walk et al 709


SCCHN, squamous cell carcinoma of the head and neck.
Figure 1. The cancer immunity cycle. Adapted from Chen DS, Mellman I.12 Oncology meets immunology: the cancer-immunity cycle. Immunity.
2013;39(1):1–10.

approved by one or more global regulatory agencies across Clinical Oncology annual meeting) as ‘‘a test that aids in the
multiple cancer indications (Table 1).4 A recent systematic benefit-risk decision-making about the use of the thera-
review and meta-analysis by Khunger et al21 examined 41 peutic product, where the difference in benefit-risk is
PD-1/PD-L1 inhibitor clinical trials and found that PD-L1 clinically meaningful.’’25 In contrast, the FDA defines a
IHC positivity in tumor cells and immune cells was companion diagnostic as an ‘‘in vitro diagnostic device or an
predictive of favorable response across all tumor types. imaging tool that provides information that is essential for
Generally, PD-L1 expression is associated with a greater the safe and effective use of a corresponding therapeutic
likelihood of benefit from anti–PD-(L)1 therapy, but, in product.’’26
contrast to traditional companion diagnostic markers such
as HER2 or ALK, patients with PD-L1–negative tumors also PD-L1 Testing
benefit from these therapies up to 20% of the time, Four separate PD-L1 assays27–30 have been developed and
depending on the specific study, therapy, and indication.22–24 approved in association with the clinical development of the
In response to these data, the FDA created a novel different FDA-approved anti–PD-(L)1 therapies and cancer
diagnostic category termed complementary diagnostic, defined indications, leading to distinct therapy-indication-test com-
(in a draft definition given at the 2016 American Society of binations (Table 3). In addition, a fifth assay based on the

Table 2. Alternative Mechanisms of Programmed Death Ligand-1 (PD-L1) Expression


Mechanism Findings Source, y
PTEN deletion, PI3K activation PTEN deletion and activation of the PI3K pathway increases PD-L1 Parsa et al,180 2007
expression in glioblastoma
ALK rearrangements ALK rearrangements have been shown to induce PD-L1 expression in Marzec et al,181 2008
anaplastic large cell lymphoma and non–small cell lung cancer Ota et al,182 2015
PD-L1 amplification/copy PD-L1 amplification/copy number alteration has been identified in multiple Roemer et al, 183 2016
number alteration tumor types, including 97% of classic Hodgkin lymphomas and more Chapuy et al,184 2016
than 50% of B-cell lymphomas
Goodman et al,185 2018
PD-L1 gene structural variations PD-L1 3 0 UTR may serve as a genetic marker for identifying cancers that Kataoka et al,186 2016
disrupting the 3 0 UTR actively evade immune surveillance and may potentially respond to
checkpoint inhibition
Stabilization of PD-L1 mRNA Oncogenic RAS signaling can upregulate tumor cell PD-L1 expression Coelho et al,187 2017
through a mechanism involving increases in PD-L1 mRNA stability via
modulation of the AU-rich element-binding protein tristetraprolin (TTP)
Abbreviations: ALK, anaplastic lymphoma kinase; mRNA, messenger RNA; UTR, untranslated region.

710 Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al
Table 3. Approved Programmed Death Ligand-1 (PD-L1) Immunohistochemistry (IHC) Assays
EMA
Assay Name Indication Drug PD-L1 Scoring Measure Indication-Specific Cutoff FDA Classification Classification
PD-L1 IHC 22C3 NSCLC, stage III, first line Pembrolizumab TPS TPS  1% PMA: companion CE mark
PharmDx NSCLC, metastatic, second line TPS TPS  1%
Gastric or GEJ adenocarcinoma, CPS CPS  1

Arch Pathol Lab Med—Vol 144, June 2020


recurrent locally advanced or
metastatic
Cervical cancer, recurrent or CPS  1
metastatic
UC, locally advanced or CPS  10
metastatic
Esophageal squamous cell CPS  10
carcinoma, recurrent, locally
advanced or metastatic
PD-L1 IHC 28-8 Nonsquamous NSCLC Nivolumab % evaluable tumor cells 1%, 5%, 10% PMA: complementary CE mark
PharmDx Melanoma exhibiting partial or 1%
complete membrane
SCCHN 1%
staining at any intensity
UC 1%
Ventana PD-L1 (SP142) UC—first line, cisplatin ineligible Atezolizumab IC 5% IC PMA: companion CE mark
assay NSCLC TC, IC 50% TC or 10% IC PMA: complementary
TNBC Atezolizumab IC 1% IC PMA: companion
Ventana PD-L1 (SP263) UC Durvalumab % tumor cells with any 25% of tumor cells exhibit PMA: complementary CE mark
assay membrane staining membrane staining; or
above background, ICþ ICP . 1% and ICþ  25%; or
ICP ¼ 1% and ICþ ¼ 100%
NSCLC—first line Pembrolizumab TC 50% NA CE mark
NSCLC—second line 1%
NSCLC—second line Nivolumab 1%, 5%, 10%
Abbreviations: CPS, combined positive score, the number of PD-L1 staining cells (tumor cells, lymphocytes, macrophages) divided by the total number of viable tumor cells, multiplied by 100; CE,
European conformity; EMA, European Medicines Agency; FDA, US Food & Drug Administration; GEJ, gastroesophageal junction; IC, the proportion of tumor area occupied by PD-L1 expressing tumor-
infiltrating immune cells of any intensity; ICþ, the percentage of tumor-associated immune cells with staining at any intensity above background; ICP, immune cells present, the percentage of tumor area
occupied by any tumor-associated immune cells; NA, Not applicable; NSCLC, non–small cell lung carcinoma; PMA, premarket approval; SCCHN, squamous cell carcinoma of the head and neck; TC,
percentage of tumor cells with PD-L1 membrane staining at any intensity above background staining as noted on the corresponding negative control; TNBC, triple-negative breast cancer; TPS, tumor
proportion score, the percentage of viable tumor cells showing partial or complete membrane staining at any intensity; UC, urothelial carcinoma.

Cancer Immunotherapy Biomarker Testing Landscape—Walk et al 711


73-10 PD-L1 clone is currently in development in associ- classification would have been made depending on which
ation with the PD-L1 inhibitor avelumab. This is a rapidly assay and cutoff combination was used. Extending the
evolving field, however, and changes to the current PD-L1 comparison to 81 additional lung cancer cases, phase 2 of
testing landscape are expected with approvals of additional the project validated the results of phase 1, with the 22C3,
immunotherapies, therapeutic indications, and assays. 28-8, and SP263 assays again demonstrating highly
Based on each combination of assay, therapy, and indica- comparable staining results and the SP-142 assay demon-
tion, the PD-L1 scoring measure and threshold for positivity strating a lower proportion of tumor cell staining.38
(ie, cutoff) can vary. For example, the companion diagnostic Additionally, the 73-10 assay was found to have a higher
assay based on the 22C3 clone for the pembrolizumab non– PD-L1 analytic sensitivity versus the other assays. Intra-
small cell lung cancer (NSCLC) first-line indication uses a reader reproducibility was high for tumor cell PD-L1 scoring
tumor proportion score with a 50% cutoff, whereas the (intraclass correlation coefficient, 0.86–0.93) but low (intra-
companion diagnostic assay based on the SP142 clone for class correlation coefficient, 0.18–0.19) for immune cell
atezolizumab in the triple-negative breast cancer indication scoring. The study also found a high concordance between
assesses tumor-infiltrating immune cells at a 1% cutoff PD-L1 scoring using glass slides versus digital images
(Table 3). The most current version of the manufacturer’s (Pearson correlation .0.96) and good agreement in
package insert and/or the interpretation guide for each assay assessing PD-L1 in cytology cell block material (intraclass
should be used for clinical testing and scoring. In addition to correlation coefficient, 0.78–0.85).
the FDA–approved assays, nonapproved PD-L1 antibody Rimm et al39 conducted a prospective, multi-institutional,
clones have been independently developed as laboratory- pathologist-based assessment of 28-8, 22c3, SP142, and
developed tests, also known as laboratory-developed assays, E1L3N PD-L1 IHC assays (the SP263 PD-L1 assay was not
per College of American Pathologists and Clinical Labora- included in this comparison). The study, which was
tory Improvement Amendments of 1988 guidelines.31,32 A sponsored by the National Comprehensive Cancer Network
2017 comprehensive review of PD-L1 laboratory-developed and funded by Bristol-Myers Squibb, found that the 22c3-
assays demonstrated variable concordance between them, based assay showed a slightly but statistically significantly
concluding that standardization is needed before PD-L1 lower staining level than the 28-8 and E1L3N assays, but the
laboratory-developed assays can be recommended for authors ultimately concluded that the 28-8, 22c3, and
routine clinical use.33 Because FDA labeling of PD-1/PD- E1L3N assays are ‘‘essentially equivalent.’’ Consistent with
L1 immunotherapies with an associated companion diag- the Blueprint project findings, the SP142 assay was
nostic requires tumors to be tested for PD-L1 as determined associated with a significantly lower mean PD-L1 expres-
by an FDA-approved test, use of PD-L1 laboratory- sion score in tumor cells. The study also found that
developed tests in these indications would be considered interreader reproducibility across assays was excellent for
off-label use of the respective therapy.34,35 tumor cell PD-L1 expression, but poor for immune cell
Based on the associated therapeutic efficacy and safety expression.
data, the FDA has designated PD-L1 as a complementary Recently, Munari et al40 conducted an interclone compar-
diagnostic for some specific PD-(L)1 inhibitor indication ison of the 22c3 and SP263 PD-L1 assays on 198 NSCLC
intended uses and as a companion diagnostic for others cases, concluding that the 2 assays are not interchangeable,
(Table 3). In addition, the FDA has demonstrated that it will with statistically significant differences seen at both the 1%
adapt drug and diagnostic labels based on emerging clinical and 50% cutoffs. At the 50% cutoff, approximately half of
and safety data. For example, in June 2018, the FDA the cases positive for SP263 would have been interpreted as
restricted the use of pembrolizumab and atezolizumab in negative for 22c3. Although the authors were not able to
patients with locally advanced or metastatic urothelial perform a direct correlation with response to anti–PD-(L)1
cancer to those whose tumors express PD-L1 and who are therapy, they concluded that these differences could lead to
not eligible for cisplatin-containing therapy. This restriction ‘‘possible underestimation of patients suitable for pembro-
was based on the finding in 2 ongoing clinical trials that PD- lizumab therapy.’’
L1–low patients treated with anti–PD-(L)1 therapy had Although these analytic comparison studies are valuable,
decreased survival compared with patients receiving che- no study to date has examined cross-assay concordance
motherapy.36 The labels of both drugs and the associated against clinical outcome endpoint measures such as
diagnostic assays were revised accordingly, thereby con- response rate, progression-free survival, or overall survival
verting PD-L1 from an optional complementary diagnostic in a common PD-(L)1 therapeutic trial. Assessment and
to a mandatory companion diagnostic for this specific validation of multiple PD-L1 assays and cutoffs across
clinical indication. multiple PD-L1 inhibitors would allow a determination of
Several studies have assessed the analytic and technical true clinical sensitivity and specificity and therefore repre-
concordance between the on-market PD-L1 IHC assays. sents a priority in the field of cancer immunotherapy.
The Blueprint Project was an industry-academic partnership PD-L1 heterogeneity, both intratumoral41 and intertu-
sponsored by the International Association for the Study of moral,42,43 has been described and represents a challenge for
Lung Cancer designed to provide information on the reliably assessing PD-L1 in the diagnostic setting. Ilie et al44
analytic comparability of the various PD-L1 assays. Phase performed a comparative study of the PD-L1 status between
1 of the project examined 39 NSCLC cases scored by 3 surgically resected specimens and matched biopsies of
pathologists across 4 PD-L1 antibody clones (22c3, 28-8, NSCLC patients. They found a discordance rate of
SP142, and SP263).37 The results demonstrated comparable approximately 50%, with the majority of the discrepancies
tumor cell staining among the 22c3, 28-8, and SP263 assays, showing PD-L1–negative biopsies and PD-L1–positive
with the SP142 assay exhibiting fewer stained tumor cells surgical resections from the same patient (75% of the
relative to the other 3 assays. Variability of immune cell discordances were due to differences in immune cell
staining was greater than for tumor cell staining across all scoring). Therefore, PD-L1–negative tumors that respond
assays, and in 37% of cases (14 of 38), a different PD-L1 to immune checkpoint inhibitors may represent heteroge-
712 Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al
Figure 2. The frequency of microsatellite instability–high (MSI-H) status of 7919 tumor and matched normal pairs across 23 tumor types. Adapted
from Cortes-Ciriano I et al.56 Nat Commun. 2017;8:15180.

neous and/or temporal expression that was not detected, or usually caused by epigenetic silencing of MLH1 via
potentially not present, at the time of initial specimen promoter methylation and are frequently associated with
testing.45 Radiation therapy, chemotherapy, and small- BRAF V600E mutations.54,55
molecule kinase inhibitors have all been shown to induce Mismatch repair deficiency was first detected in colorectal
PD-L1 expression46–48 and therefore could represent bio- cancer (CRC) but can occur in many other tumor types56,57
logical causes of pretreatment versus posttreatment PD-L1 (Figure 2), with a prevalence of 4% across all adult solid
testing discrepancy. malignancies.58 Tumors with a significant frequency of
dMMR include endometrial, gastric, small intestinal, colo-
MMR AND MSI (STATUS: FDA-APPROVED THERAPIES rectal, cervical, prostate, bile duct, liver, and thyroid
REQUIRE TESTING) carcinomas; neuroendocrine tumors; and uterine sarco-
MMR Pathobiology mas.58 In one study, endometrial, gastric, and small
The DNA MMR system recognizes and corrects insertion, intestinal cancers were more likely to have MMR defects
deletion, and base pair mismatches that occur during DNA than colon cancer.59
replication.49 Deficiencies in MMR are primarily caused by Inherited deficiencies of MMR are the cause of hereditary
inactivation of one or more of the 4 main proteins: MLH1, nonpolyposis colon cancer, also known as Lynch syndrome,
MSH2, PMS2, and MSH6. Mismatch repair deficiency a genetic syndrome with a predisposition to cancer,
(dMMR) can be inherited or acquired (sporadic) and leads especially CRC and endometrial carcinoma (52%–82% and
to accumulation of 2 main types of mutations in the DNA: 25%–60% lifetime risk, respectively). Approximately 8%–
missense mutations throughout the genome and changes in 16% of dMMR CRC cases are associated with Lynch
the length of microsatellite regions.50,51 syndrome, with the remaining cases representing sporadic
Although inherited MMR defects can be caused by CRC.60–63 Identifying cases of Lynch syndrome is critical
inactivating mutations of any of the 4 MMR genes, MLH1 given the increased cancer risk not only for the patient (ie,
and MSH2 mutations account for approximately 90% of for other tumors), but also for the patient’s family members.
cases, with mutations in PMS2 and MSH6 accounting for Therefore, it is recommended that all cases of CRC and
most of the remaining cases.52,53 Sporadic MMR defects are endometrial carcinoma undergo screening for MMR de-
Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al 713
fects.64–66 If dMMR is detected, further testing should be enabled activated T-cell immune response, are sufficient to
performed to determine if the defect is inherited or sporadic elicit antitumor immunity. Mismatch repair deficiency is
after the appropriate genetic counseling and consent is simply one mechanism that can create increased tumor
obtained. mutations, thereby increasing the probability of a neo-
antigen that can be processed and recognized by the
MSI Pathobiology adaptive immune system. For example, dMMR tumors
Microsatellites, also known as short tandem repeats or harbor a 10- to 100-fold higher rate of mutations compared
simple sequence repeats,67,68 are short repetitive DNA with pMMR tumors.74,75 High tumor mutational burden
sequences that occur commonly throughout the genome, (TMB) (see the TMB section) can be created via other defects
primarily within noncoding intergenic regions and introns. in the DNA editing machinery, such as loss of polymerase
During DNA replication, transient dissociation and subse- exonuclease function via POLE and POLD1 mutations (see
quent misaligned reassociation of the replicating DNA the POLE and POLD1 Mutations section).
strands can cause alterations in the length of these
microsatellites. These alterations are usually corrected by MMR and MSI Testing Methods
the MMR system, but in the setting of dMMR can remain There are 2 main methods of screening for MMR
uncorrected, resulting in MSI.69 Therefore, MSI is a genomic functional defects: IHC for the 4 MMR proteins MLH1,
consequence of dMMR, with the severity of instability MSH2, PMS2, and MSH6,76,77 and molecular PCR testing to
stratified by the number of affected microsatellite markers detect MSI.78–80 Each method has unique limitations and
into 3 groups: MSI high (MSI-H), MSI low, and microsat- diagnostic pitfalls, with IHC being dependent on fixation
ellite stable (see MMR and MSI Testing Methods below).69 conditions and accurate interpretation and MSI requiring
normal tissue comparison and sometimes microdissection.81
MMR, MSI, and Cancer Immunotherapy Analytically, these methods show comparable performance
Testing for dMMR and MSI has become a mandatory with an approximately 5% to 10% false-negative rate each.82
component of identifying patients most likely to respond to Mismatch repair IHC results can be falsely negative in the
immunotherapy targeting PD-(L)1 in certain indications. setting of mutations (eg, in MLH1) that result in an
The first clinical observation suggesting that MMR status antigenically intact but functionally inactive protein, and
predicted clinical response to anti–PD-1 blockade came MSI can be falsely negative in the setting of intratumoral
from a phase 2 study of the PD-1 checkpoint inhibitor heterogeneity and inadequate microdissection.83 The choice
pembrolizumab.70 Le et al70 hypothesized that the efficacy of which method to use at a given institution usually
rate of PD-1 blockade in CRC was associated with MMR depends on factors such as cost, availability, and turnaround
status and initiated a phase 2 clinical trial to evaluate time. Although using both tests will detect slightly more
pembrolizumab in patients whose tumors were dMMR or cases than either test alone, the benefit is small. In the
MMR proficient (pMMR). The results of this study showed a setting of Lynch syndrome screening, a dMMR or MSI result
dramatic difference in the objective response rate (ORR) of is typically followed by sequencing of specific MMR genes,
dMMR versus pMMR patients, with a 40% ORR in the except in the case of MLH1 loss, when BRAF V600E and/or
dMMR CRC patients versus 0% ORR in the pMMR CRC promoter methylation testing can be used to confirm
patients. Additionally, dMMR non-CRC patients, including somatic MMR deficiency.84
those with ampullary, endometrial, small bowel, and gastric Although IHC-based assessment of MMR status has been
cancers, also experienced a high rate of benefit with a 71% available for many years in the context of Lynch syndrome
ORR. screening, it is unclear what, if any, changes to the analytic
Currently, 2 cancer immunotherapy drugs are FDA and interpretative aspects of testing will be required for the
approved for dMMR tumors based on IHC or MSI application to cancer immunotherapy. Multiple IHC anti-
polymerase chain reaction (PCR) testing. In May 2017, the body clones for each of the 4 MMR proteins are available,
FDA granted accelerated approval to pembrolizumab for primarily as class 1 in vitro diagnostics, with 1 commercial
treatment of unresectable or metastatic dMMR or MSI-H MMR panel having been FDA cleared for CRC as an aid in
solid tumors that have progressed following prior treatment, the identification of probable Lynch syndrome.85,86 There are
the first cancer site–agnostic approval granted from the currently no FDA-approved MMR/dMMR or MSI assays as
FDA.71 The data for pembrolizumab were based on the companion diagnostics for cancer immunotherapy, despite
results from 5 single-arm multicenter trials (KEYNOTE-016, the fact that the FDA has approved 2 cancer immunother-
-164, -012, -028, and -158) of 149 total patients (90 CRC apies for use in dMMR/MSI-H patient subsets.
patients, 59 with 14 other cancer types) showing an ORR of The IHC determination of dMMR is based on identifying
40%, which was similar irrespective of tumor type.71 In these loss of one or more MMR proteins, indicated by absent
studies, determination of MSI-H or dMMR status for the nuclear staining in viable tumor nuclei in the presence of
majority of patients was prospectively performed via PCR unequivocal internal control cell staining. Although MMR
for MSI-H status or IHC for dMMR. The second FDA IHC staining patterns are usually consistent with the
approval related to MMR deficiency occurred when nivolu- biological function of the 4 MMR proteins, pathologists
mab was approved in July 2017 for CRC that has progressed should be aware of unusual staining patterns and diagnostic
following therapy, based on the CheckMate 142 study of 53 pitfalls.85,87 Based on data in CRC, MMR IHC assessment in
CRC patients showing an ORR of 32% in the overall biopsy samples may be preferable to surgical resection
population.72,73 specimens given that MMR protein staining loss has been
The association between dMMR response to cancer observed posttreatment.88–90 Although detection of BRAF
immunotherapy appears to be driven by the probabilistic V600E via IHC can play a role in differentiating sporadic
creation of nonself, cancer-specific antigens termed neo- CRC from Lynch syndrome,91 it does not have a role in the
antigens (see the Cancer Neoantigens section). These identification of dMMR cancers potentially responsive to
neoantigens, in the presence of a cancer immunotherapy– cancer immunotherapy.
714 Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al
Figure 3. Prevalence of somatic mutations across human cancer types. Cancer subtypes ordered from lowest average mutation prevalence (left) to
highest (right). Note extensive variation of mutation prevalence within any single cancer subtype. Abbreviations: ALL, acute lymphocytic leukemia;
AML, acute myeloid leukemia; CLL, chronic lymphocytic leukemia. Reprinted by permission from Nature Publishing Group. Alexandrov LB et al.100
Signatures of mutational processes in human cancer. Nature. 2013;500(7463):415–421.

Microsatellite instability is traditionally assessed via PCR TMB (STATUS: EMERGING)


analysis of specific microsatellite loci to determine if a Background and Role in Cancer Immunotherapy
change in length due to insertion or deletion of DNA
repeats has occurred in the tumor as compared with normal Tumor mutational burden is a measure of somatic cancer
tissue. If the same number of DNA repeats is present in the mutation prevalence typically represented as the number of
tumor and normal tissue, the result is considered microsat- mutations per megabase. Tumor mutational burden varies
ellite stability. In contrast, if the number of DNA repeats significantly across and within human cancer types, with
differs between tumor and normal, the result is considered melanoma, lung cancer, and bladder cancer having the
MSI. In 1998 the National Cancer Institute established highest mutation prevalence (Figure 3).100 Although TMB is
international criteria for MSI determination in CRC based related to dMMR and MSI, there is not complete overlap
on 5 microsatellite markers, 2 mononucleotide markers between them; most dMMR/MSI-H tumors have high TMB,
(BAT25 and BAT26), and 3 dinucleotide markers (D2S123, but not all high-TMB tumors are dMMR/MSI-H.99 For these
D5S346, and D17S250).92 Subsequently, an alternative panel reasons, TMB should be considered an independent
parameter and should not be used interchangeably with
containing 5 mononucleotide quasi-monomorphic markers
dMMR or MSI.
was proposed that demonstrated improved sensitivity
The early realization that cancer immunotherapy response
(95.8% versus 76.5%) and positive predictive value (88.5%
rates were highest among tumor types with the highest
versus 65.0%) compared with the original National Cancer
tumor mutation density led to several clinical outcome
Institute panel.93 Using either panel, a tumor is classified as
studies to further investigate this correlation. The seminal
MSI-H if 30% or more of the repeats are unstable, MSI low observation in 2014 by Snyder et al101 was that high
if less than 30% of repeats are unstable, and microsatellite mutational load in melanoma correlated with sustained
stable if no repeats are unstable.94 clinical benefit from ipilimumab or tremelimumab-based
Microsatellite instability can also be assessed via next- CTLA-4 inhibition. Importantly, the authors noted that this
generation sequencing (NGS), and a limited number of correlation was imperfect, with some high-TMB patients not
studies have shown good sensitivity and specificity using benefiting from therapy. Several subsequent studies, in-
NGS to assess MSI compared with traditional PCR cluding those of Rizvi et al102 and Hellmann et al,103,104
methods.95–97 The NGS assay must be designed to include demonstrated a similar association between TMB and
microsatellite regions, use bioinformatics algorithms that response to immunotherapy in lung cancer. The specific
can size these regions, and take into account the size immunotherapies, disease indications, and conclusions from
distribution for microsatellite regions in normal samples due these studies are listed in Table 4.
to polymerase slippage in vitro. Sensitivity of 96.4% to 100% Beyond melanoma and lung cancer, mutational load has
and specificity of 97.2% to 100% have been described for also been shown to be a predictor of response to checkpoint
both capture-based and PCR-based library preparation.98,99 blockade in a broad array of tumor types, suggesting that
Future cancer immunotherapy trials examining response high TMB could be used to identify optimally responsive
and survival rates in these discrepant cases will determine if patient subsets across all cancers. Rosenberg et al105 found
MSI-NGS provides any advantages over the traditional 5- mutational load to be an independent predictor of response
marker PCR panel. A clear advantage associated with MSI- in metastatic urothelial carcinoma patients treated with
NGS is the ability to couple the analysis with broader atezolizumab. In a retrospective analysis, Goodman et al106
genomic analyses without adding an additional test or also found that TMB was an independent predictor of
consuming additional tumor material. outcome in a study of 151 patients, multiple immunother-
Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al 715
Table 4. Lung Cancer Studies Assessing Tumor Mutational Burden (TMB) and Cancer Immunotherapy Response
Study/Drug Findings Source, y
KEYNOTE-001/pembrolizumab Higher TMB was associated with improved objective response, durable Rizvi et al,102 2015
clinical benefit, and progression-free survival
CheckMate 227/nivolumab Progression-free survival was significantly longer versus chemotherapy among Hellmann et al,103 2018
þ ipilimumab patients with high TMB, irrespective of programmed death ligand-1
expression level
CheckMate 012/nivolumab TMB was the strongest feature associated with benefit in a multivariable Hellmann et al,104 2018
þ ipilimumab analysis
CheckMate 026/nivolumab Exploratory retrospective analysis suggests improvement in objective response Peters et al,188 2017
(first line) rate and progression-free survival compared with chemotherapy in patients
with high TMB

apies, and more than 20 diverse tumor types including with mutation burden determined by WES. Examining the
breast, cervical, ovarian, prostate, and renal cancers. effect of various genomic parameters on quality, they found
Evidence to date indicates that PD-L1 status and TMB are that filtering out germline alterations and rare variants was
independent biomarkers with largely additive ability to important in determining accurate TMB measurements.
predict benefit from immune checkpoint inhibition. Car- The ability of NGS-based methods to accurately assess
bone et al107 did not observe an association between PD-L1 TMB is dependent on the size of the target region
expression and TMB in an exploratory analysis within the sequenced. Several groups have attempted to define the
CheckMate 026 phase 3 study of nivolumab as first-line minimum number of genes that can be used as the basis for
treatment of advanced NSCLC patients, finding a higher TMB, while remaining predictive of response to immuno-
response rate in those patients with a high level of both therapy. Campesato et al110 assessed the ability of their
TMB and PD-L1 expression (75%) compared with patients institutional CGP (641 genes) and the Foundation Medicine
with a high level of either biomarker alone (32% for TMB, CGP (315 genes) to estimate TMB, comparing the clinical
34% for PD-L1) or patients with low levels of both outcome prediction performance with WES-based TMB
biomarkers (16%). Hellmann et al103,104 similarly found from the melanoma and NSCLC checkpoint blockade trials
TMB and PD-L1 expression to be independent biomarkers from Snyder et al101 and Rizvi et al,102 respectively. Tumor
in the CheckMate 227 trial103 and the CheckMate 012 mutational burden estimates from either of the CGPs were
trial,104 where NSCLC patients with positive PD-L1 significantly associated with durable clinical benefit in
expression (1%) and high TMB (.median) had signifi- NSCLC patients treated with PD-1 blockade. Predictive
cantly improved response and progression-free survival accuracy of CGP-TMB for durable clinical benefit was not
rates with combination nivolumab plus ipilimumab therapy statistically different from that of TMB from WES, with
compared with patients whose tumors had only one or similar area under the curve, sensitivity, and specificity
neither variable. between the 2 CGPs and WES. Importantly, TMB predictive
accuracy was lost when smaller CGPs (,150 cancer genes)
TMB Testing were examined, causing the authors to recommend CGPs
Several challenges need to be overcome before TMB can with more than 300 cancer genes for TMB estimation. The
be readily adopted into the routine clinical environment, the inability of smaller gene panels to accurately estimate TMB
most critical being testing access, technical/analytic burden, is likely due to the sequenced region, typically in the range
and lack of standardized methods for TMB calculation and of 25 kilobases, representing too small a portion of the
reporting. genome to accurately represent overall mutational burden.
Most published studies demonstrating a correlation Another challenge preventing widespread adoption of
between high TMB and clinical response to cancer TMB as a biomarker is the lack of standardization of TMB
immunotherapies used whole-exome sequencing (WES), a calculation and reporting. For example, there is no standard
methodology not widely available or practical because of its definition of high TMB, with some studies using various
high cost and analytic burden. Targeted NGS is becoming absolute mutation thresholds102 and others using ratios of
increasingly accessible across laboratory testing environ- mutations per megabase of DNA sequenced.111 The Friends
ments, and, when coupled with optimized bioinformatics of Cancer Research, partnering with the National Cancer
pipelines, can provide an accurate and practical method for Institute, the FDA, Johns Hopkins University, Memorial
detecting the sequence changes used to calculate TMB. For Sloan Kettering Cancer Center, and multiple pharmaceutical
example, NGS-based comprehensive gene panels (CGPs) and diagnostic companies, hosted a TMB harmonization
sequence specific sets of genes (typically 300–600) related to working group meeting in May 2018 to begin the process of
a focused disease or phenotype. These panels represent a addressing these challenges.112 Several opportunities for
more routinely feasible option for assessing TMB in the TMB technical harmonization were provided, such as
routine laboratory environment because of their lower cost, agreement on analytic parameters for TMB calculation,
faster turnaround time, and lower analytic burden compared generation of a universal reference standard as an alignment
with WES. Several studies have found NGS-based TMB tool, and agreement on statistical approaches that will lead
assessment to be an accurate surrogate of TMB by WES. to consistent TMB calculation and clinical interpretation.
Rizvi et al108 studied TMB assessment by targeted NGS Tumor mutational burden testing currently requires access
versus WES, finding a strong correlation between the 2 to tumor tissue, but noninvasive blood-based means of
methods. Chalmers et al109 found that TMB from a 1.1- estimating TMB are being explored. Gandara et al113
megabase targeted CGP was strongly correlated (R2 ¼ 0.74) developed a novel blood-based assay to measure TMB
716 Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al
based on single-nucleotide variants from 394 genes from response can be generated.124 In this way, dMMR, MSI-H,
cell-free DNA in patient plasma. Using blood samples from and TMB can all be thought of as surrogate measures of
the atezolizumab POPLAR (211 patients) and OAK (583 immunogenic neoantigens, which may be the actual targets
patients) trials in second-line NSCLC, the blood-based TMB of cytotoxic T cells in clinical cases of immunotherapy
assay was predictive of clinical benefit independent of PD- response. In both melanoma and NSCLC patients, higher
L1 expression. Prospective validation of this blood-based neoantigen burden is associated with improved clinical
assay as an alternative to tissue-based TMB would improve responses to immune checkpoint blockade therapy.125
the routine implementation of mutational burden assess- Based on these hypotheses and early observations, there is
ment in clinical practice. great interest in using tumor sequencing and bioinformatic
prediction algorithms to directly identify neoantigens that
POLE AND POLD1 MUTATIONS (STATUS: EMERGING) can be used therapeutically in cancer vaccines126 and
Background and Biology diagnostically as more direct predictive biomarkers of cancer
immunotherapy efficacy. Experimental data exploring the
Polymerase d (POLD1 gene) and polymerase e (POLE frequency, identity, and uniqueness of cancer neoantigens
gene) are DNA polymerases involved in DNA replication have revealed new information that will guide the clinical
during the S phase of the cell cycle.114 These enzymes translation of this potentially important immunotherapy
additionally have DNA proofreading and repair functional-
biomarker.
ity via an exonuclease domain that allows excision and
Although human T cells have been shown to react to both
replacement of incorrect bases and continuation of DNA
major histocompatibility complex class I–restricted and
replication. Mutations of the exonuclease domain disrupt
major histocompatibility complex class II–restricted neo-
the proofreading function and lead to an accumulation of
antigens in a large fraction of malignancies,127–130 only a
mutations elsewhere in the genome, often referred to the
small percentage of tumor mutations actually lead to the
ultramutator phenotype.114
formation of neoantigens that are recognized by host T
POLE mutations associated with the ultramutator pheno-
cells.126,131–133 Additionally, it appears that immunoreactive
type have been described in many different cancer types,
cancer neoantigens can arise from mutations in any gene
including about 3% of CRCs and 7% to 10% of endometrial
cancers.115–118 These tumors have thousands of mutations across the genome, and are not more common in driver
across the genome, an inflamed tumor microenvironment, oncogenes related to tumorigenesis.126,129,134 Steven Rosen-
and upregulated PD-L1, and are associated with a better berg, MD (National Cancer Institute, Bethesda, Maryland),
prognosis, similar to dMMR tumors. POLE mutations are pioneered a tandem minigene approach to identify the
predominantly found in microsatellite-stable/pMMR tu- precise neoantigens recognized by T cells from patients
mors, but one study identified them in MSI cases and treated with adoptive cell transfer.135,136 In melanoma,
found that they account for some unexplained Lynch neoantigens were identified in 29 of 31 patients (94%),
syndrome phenotypes.119 POLD1 mutations appear to be with each of the identified neoantigens being unique to the
less prevalent than POLE mutations and occur more autologous patient.135 In common gastrointestinal cancers,
commonly in dMMR tumors, but can also lead to the neoantigen-reactive T cells were identified in 62 of 75
ultramutator phenotype.120,121 patients (83%), with 99% of the neoantigenic determinants
unique to each autologous patient.136 Immunogenic tumor
Implications for Cancer Immunotherapy and Testing antigens associated with adoptive cell transfer complete
Given the high mutation rate, inflamed tumor microen- regressions have been identified in metastatic breast
vironment, and upregulation of immune checkpoints cancer137 and human papillomavirus–associated metastatic
associated with POLE-mutated cancers, there is strong cervical cancer.138
scientific rationale to assess the efficacy of cancer immuno- Although this area is promising, several challenges would
therapy in this setting. If effective, it would bring clinical need to be overcome before neoantigen prediction or
benefit to 1% to 3% of colon cancer patients not currently neoantigen load could become part of routine cancer
addressed by cancer immunotherapy. Targeted NGS or immunotherapy testing. These include lack of a complete
allele-specific PCR testing of POLE mutations could focus on understanding of the underlying immunobiology, the low
the 3 recurrent mutations (P286R/H/S, V411L, and S459F) frequency and apparent uniqueness of cancer neoantigens,
that represent 90% of the identified exonuclease domain and a lack of suitable bioinformatics and laboratory methods
mutations.122 In addition to POLD1/POLE and MMR, there is that can both provide accurate prediction and be imple-
a strong rationale to evaluate deficiencies in other DNA mented into routine practice.
repair enzymes and mechanisms, such as O6-methylgua-
nine-DNA methyltransferase (MGMT), homologous recom- TIL ASSESSMENT AND MULTIPLEXED IMMUNE
bination, nucleotide excision repair, and base excision PHENOTYPING (STATUS: EMERGING)
repair, as potential biomarkers of immunotherapy response. Pathologists have long recognized the presence of TILs in
Several translational and clinical studies exploring these diagnostic specimens, and many studies have explored their
DNA damage repair pathways in this context are currently role as biomarkers. In 2011, Mahmoud et al139 was the first
underway.123 to identify TILs as a prognostic factor in breast cancer,
showing that tumor-infiltrating CD8þ lymphocytic density is
CANCER NEOANTIGENS (STATUS: EARLY EMERGING) significantly associated with improved clinical outcome
The correlation of dMMR, MSI-H, TMB, and clinical independent of standard factors such as tumor grade,
benefit from immune checkpoint blockade is thought to lymph node stage, and HER2 status. In a study of stage I
represent a probabilistic relationship in which higher to IV CRCs, Mlecnik et al140 found that an assessment of
mutational burden within a tumor increases the likelihood CD8þ T cells in the tumor center and invasive margin was a
of a neoantigen to which an effective T-cell immune novel indicator of recurrence beyond TNM staging. Rakaee
Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al 717
et al141 developed and validated a hematoxylin-eosin–based immune subsets,147,152–154 some studies have begun to
TIL scoring model, demonstrating that high stromal TIL correlate the phenotype and cellular interactions of immune
level is an independent measure of prognosis in stage I to III cells in the tumor microenvironment with immunotherapy
NSCLC patients. Smyrk et al142 proposed TILs as a outcome data as a potentially more powerful predictor of
screening criterion for selecting which CRC samples require response. Giraldo et al155 found in the setting of Merkel cell
MSI testing, demonstrating a 93% sensitivity and 62% carcinoma that the number of pretreatment PD-1þ immune
specificity for MSI-H status with a TIL cutoff of 5. Assuming cells in proximity to (within 15 lm of) a PD-L1þ immune or
a 12% MSI-H rate, they estimated that TIL assessment tumor cell was associated with pembrolizumab response,
could reduce the number of colon cancer samples referred to which was not the case with CD8/PD-L1 proximity.
MSI testing by more than 50% while still identifying 93% of Similarly, Johnson et al156 found that a PD-1/PD-L1
MSI-H cases. interaction score, but not PD-L1 alone, was associated with
Tumor-infiltrating lymphocytes indicate an inflamed anti–PD-1 response in metastatic melanoma.
tumor microenvironment, and in the era of cancer Taken together, these studies demonstrate the potential of
immunotherapy have been assessed as a predictive bio- TILs as an important cancer immunotherapy biomarker.
marker of response. Tumeh et al19 found a significantly However, adoption of TIL assessment in any of its forms
higher CD8þ T-cell density in the baseline biopsies of into routine clinical practice will require robust methods and
melanoma patients responding to pembrolizumab com- tools for interpretation. Addressing the need for a stan-
pared with the progression group, both at the invasive dardized, reproducible approach to the histopathologic
margin and at the tumor center. However, there was no assessment and quantification of TILs in routine practice,
CD8-positive cutoff value clearly separating responders Hendry et al,157,158 on behalf of the International Immuno-
from nonresponders. Chen et al143 also found an increase oncology Biomarkers Working Group, have proposed a
in the density of CD8þ, CD3þ, and CD45ROþ T cells in hematoxylin-eosin–based methodology for assessing TILs in
pretreatment melanoma samples of CTLA-4 blockade solid tumors for clinical validity and utility assessment. In
responders compared with nonresponders, but, similar to addition, the International TILs Working Group has
Tumeh et al,19 could not establish a clear separation published methodologic recommendations on the evalua-
between the 2 groups. However, their study also included tion of TILs in breast cancer, focusing on the stromal (versus
a separate analysis of anti–PD-1 treated patients, in which intratumoral) compartment.159 Beyond manual human
on-treatment biopsies showed highly statistically significant, interpretation of TILs, computational image analysis–based
largely nonoverlapping expression differences between TIL scoring approaches have been developed, including
responders and nonresponders for CD8, CD4, and CD3 T- traditional object-oriented image-segmentation methods
cell subsets in addition to the immune checkpoint markers and more advanced machine learning–based classification
PD-L1, PD-1, and LAG3.143 These changes were seen as algorithms that rely on extensive training sets but are able to
early as after 2 or 3 doses in responding patients. Also morphologically identify cellular subsets.160
observed in the early on-treatment biopsies was an
enrichment of CD8þ T cells in the tumor center versus the TRANSCRIPTIONAL SIGNATURES OF IMMUNE
tumor margin in responders compared with nonresponders, RESPONSIVENESS (STATUS: EMERGING)
suggesting therapy-induced tumor infiltration.
Beyond the assessment of TILs by hematoxylin-eosin RNA-based gene expression studies have been used to
morphology and single-marker IHC, the development of identify specific transcriptional patterns and signatures in
advanced in situ multiplexing methods has enabled more tumors and the tumor microenvironment that may help
comprehensive immunophenotyping of tumors and the host elucidate details of immune physiology underlying immu-
microenvironment, including the ability to visualize the notherapy benefit. Summarized here are selected studies
colocalization of multiple immune-related markers.144 A exploring gene expression signatures that appear to play an
detailed overview of the currently available IHC multiplex- important role in the prediction of cancer immunotherapy
ing methods and technologies is beyond the scope of this response.
article, but can be found in recently published reviews.145,146 Taube et al161,162 found that immune infiltrates at the
The historic challenge of creating a multiplexed indirect IHC interface of PD-L1–positive melanoma cells demonstrate a
assay that avoids multiple species of primary antibodies and CD8þ T cell–T helper 1 cytokine mRNA signature charac-
cross-reactivity between same-species primary antibodies terized by IFN-c expression, which was absent in PD-L1–
has largely been solved by elegant chemical and heat negative melanomas. They hypothesized that secretion of
deactivation reactions.145 Chromogenic multiplexing allows the IFN-c cytokine not only triggers antitumor effects, but
light microscopy visualization, with novel methods such as simultaneously induces PD-L1 as part of an adaptive
multiplexed immunohistochemical consecutive staining on negative-feedback immune regulatory mechanism. Since
a single slide and reagents such as narrow-band tyramine- this seminal discovery, the adaptive expression of PD-L1
conjugated dyes demonstrating the feasibility of medium- to has been observed in other tumor types, including Merkel
high-dimensional analysis.147,148 Fluorescence technologies cell carcinoma,163 NSCLC,164 and breast cancer.165,166 Acti-
enable even higher-order multiplexing, with up to 61 vation of the IFN-c signaling pathway is now recognized as
individual analytes demonstrated with an iterative staining, a necessary component of successful anticancer T-cell
imaging, and chemical inactivation cycle approach.149 The immunity, an association reinforced by the discovery of
demonstration of fully automated fluorescence multiplexing immunotherapy resistance mutations in Janus kinase 1
using tyramide-based covalent detection methods offers the (JAK1) and Janus kinase 2 (JAK2) (see the Biomarkers of
promise of these methods entering routine anatomic Resistance to Cancer Immunotherapy section) that prevent
pathology practice.150,151 upregulation of IFN-c target genes.167
Although initial studies using these novel techniques Ribas et al168 developed 2 gene signatures, IFN-c 10-gene
focused on the characterization of specific intratumoral and expanded-immune 28-gene, both of which were
718 Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al
statistically associated with overall response rate and ing, angiogenesis, wound healing, cell adhesion, and
progression-free survival in the context of melanoma treated mesenchymal transition. Activation of the innate anti–PD-
with pembrolizumab. In NSCLC, an 8-gene T-effector and 1 resistance signature was also demonstrated in lung
interferon-c gene signature was associated with improved adenocarcinoma, colonic adenocarcinoma, renal clear cell
overall survival with atezolizumab in the POPLAR phase 2 carcinoma, and pancreatic adenocarcinoma.175
trial.169 This result supports the hypothesis that preexisting Acknowledging that the outcome of cancer-immune
immunity is a predictor of benefit from checkpoint blockade. interactions is based on multiple independent parameters
In addition to having a possible role as a clinical that can each vary between patients, Blank et al176 have
biomarker, gene expression signatures drive much of the proposed a ‘‘cancer immunogram’’ visualization framework
ongoing translational research into novel mechanisms of as a more comprehensive assessment of immune respon-
cancer immunotherapy response and resistance that could siveness and mechanisms of resistance. The immunogram
inform rational combination strategies. contains 7 initial parameter classes: tumor foreignness,
general immune status, immune cell infiltration, absence of
BIOMARKERS OF RESISTANCE TO CANCER checkpoints, absence of soluble inhibitors, absence of
IMMUNOTHERAPY (STATUS: EMERGING) inhibitory tumor metabolism, and tumor sensitivity to
Although cancer immunotherapy delivers unprecedented immune effectors. Resistance to immunotherapy is predict-
levels of durable clinical benefit to select patient subsets, ed when any of the 7 parameters is unfavorable, and only
most patients initially treated with these therapies do not overcome with the appropriate targeting of the relevant
respond (primary resistance) and of those who do respond, defect. Should this immunogram or a similar tool prove to
some experience only short-lived benefit with eventual be valuable in the selection of responsive patients, its clinical
tumor relapse (acquired or secondary resistance). This adoption will require the multifactorial integration of tumor
section will briefly summarize some of the latest transla- genomic, immunohistochemical, and peripheral blood data
tional research into the mechanisms of immunotherapy by anatomic and molecular pathologists.
resistance.
One type of cancer immunotherapy resistance is immune MICROBIOME (STATUS: EARLY EMERGING)
evasion resulting from tumor mutations that cause failures The human microbiome consists of the trillions of
in immune-mediated destruction and detection of cancer microorganisms that colonize the skin, mucosal surfaces,
cells. For example, Zaretsky et al167 identified JAK1 and and gastrointestinal tract. The role of the microbiome and its
JAK2 mutations in 2 melanoma patients who initially associated impact on human health and response to cancer
responded to pembrolizumab anti–PD-1 therapy but later therapy has been proposed and explored in the past, but
progressed. Mutations in JAK1/2 lead to abrogation of IFN- until recently has been controversial. Recent conclusive data
c signaling and its antiproliferative effect on tumor cells. supporting the role of the microbiome as a biomarker of
Shin et al170 identified JAK 1/2 mutations as the cause of response to cancer immunotherapy have reignited scientific
primary resistance to anti–PD-1 therapy in dMMR colon interest in this field and triggered significant clinical
cancer and melanoma patients, demonstrating that these validation efforts.
mutations can cause both primary and secondary forms of Routy et al177 established a correlation between gut
immunotherapy resistance. microbiota and response to checkpoint inhibition, finding
Mutations causing impairment of the HLA class 1 antigen a significant shortening of OS and progression-free survival
processing and presentation machinery appears to be in NSCLC and urothelial carcinoma patients treated with
another mechanism of resistance to cancer immunotherapy broad-spectrum antibiotics prior to anti–PD1/PD-L1 ther-
agents. Gettinger et al171 identified homozygous loss of b2- apy. Additionally, the authors found that gut microbiota
microglobulin (B2M), critical to proper functioning of the diversity, as measured by shotgun-sequencing gene count
HLA class 1 complex, in 1 of 14 lung cancer patients with or metagenomic species levels, correlated with clinical
acquired resistance to immune checkpoint inhibition. response. They found enrichment of specific bacterial genera
Zaretsky et al167 also identified a single patient with B2M in responders versus nonresponders, with Akkermansia
loss in their study of melanoma patients who progressed muciniphila having the most significant association with
after initially responding to immune checkpoint therapy. favorable clinical outcome. They concluded that the gut
Defects in several traditional cell signaling pathways have microbiome markedly influences PD-L1 blockade outcome
also been correlated with immunotherapy resistance, and hypothesized that certain bacteria such as A muciniphila
suggesting that many of these pathways can impact immune may reinforce intestinal barrier integrity, reduce systemic
regulation. Some pathways and genomic changes that have inflammation, and improve immunosurveillance.
been associated with negative immune regulation include b- Matson et al178 used 16S ribosomal RNA gene amplicon
catenin activation (Spranger et al172), PTEN loss (Peng et sequencing and species-specific quantitative PCR to study
al173), and EGFR mutations and ALK rearrangements stool samples from 42 metastatic melanoma patients
(Gainor et al174). The apparent connectivity between these receiving immune checkpoint therapy, finding 8 bacterial
pathways and immune activation/suppression provides a species more abundant in responders and 2 species more
strong rationale for combining targeted therapies and abundant in nonresponders. Supporting the findings of
immunotherapies. Routy et al,177 they identified A muciniphila in 4 patients, all
Melanomas demonstrating primary resistance to check- of whom were responders. They propose that multiple
point inhibitors were found to display a specific transcrip- specific bacteria may contribute to improved antitumor
tional signature referred to as innate anti–PD-1 immunity, and that the optimal biomarker may be a ratio of
resistance.175 Hugo et al175 found that this transcriptional beneficial bacteria that activate the immune system to
signature is characterized by increased expression of genes nonbeneficial bacteria that negatively regulate immune
involved in the regulation of extracellular matrix remodel- responses. The authors conclude that the commensal
Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al 719
microbiota may be a useful biomarker to predict response to The biomarkers reviewed here represent only a small
checkpoint blockade therapy, with patient responder- proportion of the genomic, proteomic, and immunologic
associated bacteria having distinct local and systemic effects parameters being explored in this dynamic and evolving
on innate and adaptive immunity. field. The breadth of exploration, in both the therapeutic and
Gopalakrishnan et al179 prospectively collected micro- diagnostic realms, is an indicator of our still nascent
biome samples from metastatic melanoma patients prior to understanding of the mechanisms underlying immune-
anti–PD-1 therapy. Within-sample gut microbiome diversity mediated cancer destruction. Adherence to the translational
was found to be significantly higher in responders versus medicine philosophy, whereby laboratory research leads to
nonresponders, with greater diversity also correlating with novel therapeutic and diagnostic hypotheses that are then
prolonged progression-free survival. Specific compositional clinically tested to inform subsequent scientific investiga-
differences associated with anti–PD-1 therapy were identi- tions, will continue to drive cancer immunotherapy pro-
fied, with Clostridiales order, Ruminococcaceae family, and gress, ultimately leading to better outcomes for patients.
Faecalibacterium genus enriched in responders and Bacter- As the field of cancer immunotherapy continues to
oidales order enriched in nonresponders. Exploring poten- develop, pathologists bear an important responsibility to
tial mechanisms underlying these associations, the authors lead and support studies that explore and validate novel
found a statistically significant correlation between CD8þ T- biomarkers. The current dynamism of this field represents
cell infiltrates in the tumor and abundance of Faecalibacte- an opportunity for pathology to establish itself as the
rium genus, Ruminococcaceae family, and Clostridiales immunotherapy biomarker center of excellence in clinical
order in the gut, suggesting enhanced systemic and medicine, ensuring the accurate, standardized, and timely
antitumor responses mediated by increased antigen pre- implementation of immunotherapy diagnostics through
sentation and improved effector T-cell function. Finally, education, training, and collaboration.
germ-free mice transplanted with stool from responders to
anti–PD-1 therapy had higher CD8þ T-cell density and The authors would like to gratefully acknowledge Molly Hansen,
improved response to immune checkpoint blockade com- CT(ASCP), for her valuable assistance during the development of
pared with those transplanted with stool from nonrespond- this manuscript.
ers, suggesting a causal link between a favorable gut References
microbiome and immune checkpoint therapy. 1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell.
Collectively, these studies provide compelling evidence 2011;144(5):646–674.
2. Callahan MK, Postow MA, Wolchok JD. Targeting T cell co-receptors for
that the diversity and composition of the gut microbiome cancer therapy. Immunity. 2016;44(5):1069–1078.
could serve as a biomarker of cancer immunotherapy 3. Hodi FS, Ballinger M, Lyons B, et al. Immune-Modified Response Evaluation
response. Although additional validation via clinical trials Criteria In Solid Tumors (imRECIST): refining guidelines to assess the clinical
benefit of cancer immunotherapy. J Clin Oncol. 2018;36(9):850–858.
is necessary, the data also suggest that modulation of the 4. US Food & Drug Administration. Hematology/oncology (cancer) approvals
gut microbiome may have therapeutic potential for cancer & safety notifications. https://www.fda.gov/drugs/informationondrugs/
patients receiving immune checkpoint blockade. approveddrugs/ucm279174.htm. Accessed August 11, 2018.
5. Tang J, Shalabi A, Hubbard-Lucey VM. Comprehensive analysis of the
clinical immuno-oncology landscape. Ann Oncol. 2018;29(1):84–91.
CONCLUDING REMARKS 6. Carretero-Gonzalez A, Lora D, Ghanem I, et al. Analysis of response rate
The field of cancer immunotherapy is still young and in with ANTI PD1/PD-L1 monoclonal antibodies in advanced solid tumors: a meta-
analysis of randomized clinical trials. Oncotarget. 2018;9(9):8706–8715.
evolution, but has already transformed clinical oncology by 7. Kourie HR, Klastersky J. Immune checkpoint inhibitors side effects and
providing durable long-term survival benefit to previously management. Immunotherapy. 2016;8(7):799–807.
untreatable cancer patients. Because our ability to accurately 8. Dranoff G. Cytokines in cancer pathogenesis and cancer therapy. Nat Rev
identify the minority of patients who currently derive benefit Cancer. 2004;4(1):11–22.
9. McCarthy EF. The toxins of William B. Coley and the treatment of bone and
from these therapies remains very limited, there is a critical soft-tissue sarcomas. Iowa Orthop J. 2006;26:154–158.
need to research, develop, and translate biomarkers 10. Prehn RT, Main JM. Immunity to methylcholanthrene-induced sarcomas. J
predictive of response. Natl Cancer Inst. 1957;18(6):769–778.
11. Old LJ, Clarke DA, Benacerraf B. Effect of bacillus Calmette-Guérin
From the landscape of cancer immunotherapy biomarkers infection on transplanted tumours in the mouse. Nature. 1959;184(suppl 5):291–
reviewed in this article, only PD-L1, MMR, and MSI testing 292.
can be considered validated and routine because each is 12. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity
associated with FDA-approved therapies. Tumor mutational cycle. Immunity. 2013;39(1):1–10.
13. Pardoll DM. The blockade of immune checkpoints in cancer immuno-
burden remains emerging, with issues of sensitivity/speci- therapy. Nat Rev Cancer. 2012;12(4):252–264.
ficity and standardization yet to be resolved. Polymerase d 14. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity
and e mutations are rare events but may help clarify which by CTLA-4 blockade. Science. 1996;271(5256):1734–1736.
15. US Food & Drug Administration. YERVOY (ipilimumab) injection, for
pMMR patients derive benefit. Assessment of immunogenic intravenous use. https://www.accessdata.fda.gov/drugsatfda_docs/label/2015/
cancer neoantigens appears to be critical to understanding 125377s073lbl.pdf. Accessed October 15, 2018.
the extent of tumor foreignness, but their detection and 16. Nobel Media AB. The Nobel Prize in Physiology or Medicine 2018. https://
identification are limited by current bioinformatic and www.nobelprize.org/prizes/medicine/2018/summary. Accessed October 15,
2018.
genomic technology. Tumor-infiltrating lymphocyte assess- 17. Ribas A. Tumor immunotherapy directed at PD-1. N Engl J Med. 2012;
ment, especially enabled by multiplexed characterization of 366(26):2517–2519.
immune cell subsets, is a promising emerging biomarker 18. Spranger S, Gajewski T. Rational combinations of immunotherapeutics
that target discrete pathways. J Immunother Cancer. 2013;1:16.
that sits squarely in the domain of the anatomic pathologist. 19. Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces
Finally, transcriptional signatures of immune responsive- responses by inhibiting adaptive immune resistance. Nature. 2014;515(7528):
ness, biomarkers of cancer immunotherapy resistance, and 568–571.
the microbiome are all promising early emerging biomark- 20. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade.
Science. 2018;359(6382):1350–1355.
ers of immunotherapy response that require additional 21. Khunger M, Hernandez AV, Pasupuleti V, et al. Programmed cell death 1
scientific, analytic, and clinical validation. (PD-1) ligand (PD-L1) expression in solid tumors as a predictive biomarker of

720 Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al
benefit from PD-1/PD-L1 axis inhibitors: a systematic review and meta-analysis. 46. Akbay EA, Koyama S, Carretero J, et al. Activation of the PD-1 pathway
JCO Precis Oncol. 2017;1:1–15. contributes to immune escape in EGFR-driven lung tumors. Cancer Discov. 2013;
22. Mahoney KM, Atkins MB. Prognostic and predictive markers for the new 3(12):1355–1363.
immunotherapies. Oncology (Williston Park). 2014;28(suppl 3):39–48. 47. Leduc C, Adam J, Louvet E, et al. TPF induction chemotherapy increases
23. Weber JS, D’Angelo SP, Minor D, et al. Nivolumab versus chemotherapy PD-L1 expression in tumour cells and immune cells in head and neck squamous
in patients with advanced melanoma who progressed after anti-CTLA-4 treatment cell carcinoma. ESMO Open. 2018;3(1):e000257.
(CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet 48. Lim YJ, Koh J, Kim S, et al. Chemoradiation-induced alteration of
Oncol. 2015;16(4):375–384. programmed death-ligand 1 and CD8(þ) tumor-infiltrating lymphocytes identified
24. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated patients with poor prognosis in rectal cancer: a matched comparison analysis. Int
melanoma without BRAF mutation. N Engl J Med. 2015;372(4):320–330. J Radiat Oncol Biol Phys. 2017;99(5):1216–1224.
25. Scheerens H, Malong A, Bassett K, et al. Current status of companion and 49. Jiricny J. The multifaceted mismatch-repair system. Nat Rev Mol Cell Biol.
complementary diagnostics: strategic considerations for development and 2006;7(5):335–346.
launch. Clin Transl Sci. 2017;10(2):84–92. 50. Hewish M, Lord CJ, Martin SA, Cunningham D, Ashworth A. Mismatch
26. US Food & Drug Administration. List of cleared or approved companion repair deficient colorectal cancer in the era of personalized treatment. Nat Rev
diagnostic devices (in vitro and imaging tools). https://www.fda.gov/ Clin Oncol. 2010;7:197.
MedicalDevices/ProductsandMedicalProcedures/InVitroDiagnostics/ 51. Vilar E, Gruber SB. Microsatellite instability in colorectal cancer—the
ucm301431.htm. Accessed July 10, 2018. stable evidence. Nat Rev Clin Oncol. 2010;7:153.
27. US Food & Drug Administration. Dako PD-L1 IHC 22C3 pharmDx. https:// 52. Peltomaki P, Vasen H. Mutations associated with HNPCC predisposition—
www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfpma/pma.cfm?id¼P150013S009. update of ICG-HNPCC/INSiGHT mutation database. Dis Markers. 2004;20(4–5):
Accessed July 10, 2018. 269–276.
28. US Food & Drug Administration. PD-L1 IHC 28-8 PHARMDX. https:// 53. Buza N, Ziai J, Hui P. Mismatch repair deficiency testing in clinical
www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfpma/pma.cfm?id¼P150025S003. practice. Expert Rev Mol Diagn. 2016;16(5):591–604.
Accessed August 11, 2018. 54. Boissiere-Michot F, Frugier H, Ho-Pun-Cheung A, Lopez-Crapez E,
29. US Food & Drug Administration. VENTANA PD-L1 (SP263) assay. https:// Duffour J, Bibeau F. Immunohistochemical staining for p16 and BRAFV600E is
www.accessdata.fda.gov/cdrh_docs/pdf16/p160046c.pdf. Accessed July 10, useful to distinguish between sporadic and hereditary (Lynch syndrome-related)
2018. microsatellite instable colorectal carcinomas. Virchows Arch. 2016;469(2):135–
30. US Food & Drug Administration. VENTANA PD-L1(SP142) CDX assay. 144.
h t t p s : / / w w w. a c c e s s d a t a . f d a . g o v / s c r i p t s / c d r h / c f d o c s / c f p m a / p m a . 55. Luey N, Toon CW, Sioson L, et al. A further investigation of combined
cfm?id¼P160002S006. Accessed July 10, 2018. mismatch repair and BRAFV600E mutation specific immunohistochemistry as a
31. Ilié M, Ngo-Mai M, Long-Mira E, et al. Using 22C3 anti-PD-L1 antibody predictor of overall survival in colorectal carcinoma. PLoS One. 2014;9(8):
concentrate on biopsy and cytology samples from non-small cell lung cancer e106105.
patients. J Vis Exp. 2018(139). doi:10.3791/58082 56. Cortes-Ciriano I, Lee S, Park WY, Kim TM, Park PJ. A molecular portrait of
32. Munari E, Zamboni G, Lunardi G, et al. PD-L1 expression in non-small microsatellite instability across multiple cancers. Nat Commun. 2017;8:15180.
cell lung cancer: evaluation of the diagnostic accuracy of a laboratory developed 57. Van de Water NS, Jeevaratnam P, Browett PJ, Stewart SM, Lane MR, Jass
test using clone E1L3N in comparison with 22C3 and SP263 assays [published JR. Direct mutational analysis in a family with hereditary non-polyposis
colorectal cancer. Aust N Z J Med. 1994;24(6):682–686.
online May 21, 2019]. Hum Pathol. 2019;90:54–59. doi:10.1016/j.humpath.
58. Bonneville R, Krook MA, Kautto EA, et al. Landscape of microsatellite
2019.05.003
instability across 39 cancer types. JCO Precis Oncol. 2017(1):1–15.
33. Buttner R, Gosney JR, Skov BG, et al. Programmed death-ligand 1
59. Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts
immunohistochemistry testing: a review of analytical assays and clinical
response of solid tumors to PD-1 blockade. Science. 2017;357(6349):409–413.
implementation in non-small-cell lung cancer. J Clin Oncol. 2017;35(34):3867–
60. Aaltonen LA, Salovaara R, Kristo P, et al. Incidence of hereditary
3876.
nonpolyposis colorectal cancer and the feasibility of molecular screening for the
34. US Food & Drug Administration. In vitro companion diagnostic devices:
disease. N Engl J Med. 1998;338(21):1481–1487.
guidance for industry and Food and Drug Administration staff. https://www.fda.
61. Hampel H, Frankel WL, Martin E, et al. Screening for the Lynch syndrome
gov/media/81309/download. Accessed June 4, 2019.
(hereditary nonpolyposis colorectal cancer). N Engl J Med. 2005;352(18):1851–
35. US Food & Drug Administration. Principles for codevelopment of an in
1860.
vitro companion diagnostic device with a therapeutic product: draft guidance for
62. Poynter JN, Siegmund KD, Weisenberger DJ, et al. Molecular character-
industry and Food and Drug Administration staff. https://www.fda.gov/media/
ization of MSI-H colorectal cancer by MLHI promoter methylation, immunohis-
99030/download. Accessed June 4, 2019. tochemistry, and mismatch repair germline mutation screening. Cancer Epidemiol
36. US Food & Drug Administration. FDA alerts health care professionals and Biomarkers Prev. 2008;17(11):3208–3215.
oncology clinical investigators about an efficacy issue identified in clinical trials 63. Ward RL, Hicks S, Hawkins NJ. Population-based molecular screening for
for some patients taking Keytruda (pembrolizumab) or Tecentriq (atezolizumab) Lynch syndrome: implications for personalized medicine. J Clin Oncol. 2013;
as monotherapy to treat urothelial cancer with low expression of PD-L1. https:// 31(20):2554–2562.
www.fda.gov/Drugs/DrugSafety/ucm608075.htm. Accessed July 14, 2018. 64. National Comprehensive Cancer Network. NCCN clinical practice
37. Hirsch FR, McElhinny A, Stanforth D, et al. PD-L1 immunohistochemistry guidelines in oncology: colon cancer version 2.2018. https://www.nccn.org.
assays for lung cancer: results from phase 1 of the Blueprint PD-L1 IHC Assay Accessed July 15, 2018.
Comparison Project. J Thorac Oncol. 2017;12(2):208–222. 65. National Comprehensive Cancer Network. NCCN clinical practice
38. Tsao MS, Kerr KM, Kockx M, et al. PD-L1 immunohistochemistry guidelines in oncology: uterine neoplasms version 2.2018. https://www.nccn.
comparability study in real-life clinical samples: results of Blueprint phase 2 org. Accessed July 15, 2018.
project. J Thorac Oncol. 2018;13(9):1302–1311. 66. National Comprehensive Cancer Network. NCCN clinical practice
39. Rimm DL, Han G, Taube JM, et al. A prospective, multi-institutional, guidelines in oncology: genetic/familial high-risk assessment: colorectal version
pathologist-based assessment of 4 immunohistochemistry assays for PD-L1 1.2018. https://www.nccn.org. Accessed July 15, 2018.
expression in non-small cell lung cancer. JAMA Oncol. 2017;3(8):1051–1058. 67. Ellegren H. Microsatellites: simple sequences with complex evolution. Nat
40. Munari E, Rossi G, Zamboni G, et al. PD-L1 assays 22C3 and SP263 are Rev Genet. 2004;5(6):435–445.
not interchangeable in non-small cell lung cancer when considering clinically 68. Li YC, Korol AB, Fahima T, Nevo E. Microsatellites within genes: structure,
relevant cutoffs: an interclone evaluation by differently trained pathologists. Am J function, and evolution. Mol Biol Evol. 2004;21(6):991–1007.
Surg Pathol. 2018;42(10):1384–1389. 69. Imai K, Yamamoto H. Carcinogenesis and microsatellite instability: the
41. Haragan A, Field JK, Davies MPA, Escriu C, Gruver A, Gosney JR. interrelationship between genetics and epigenetics. Carcinogenesis. 2008;29(4):
Heterogeneity of PD-L1 expression in non-small cell lung cancer: implications 673–680.
for specimen sampling in predicting treatment response. Lung Cancer. 2019;134: 70. Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch-
79–84. repair deficiency. N Engl J Med. 2015;372(26):2509–2520.
42. Liu Y, Dong Z, Jiang T, et al. Heterogeneity of PD-L1 Expression among the 71. US Food & Drug Administration. FDA grants accelerated approval to
different histological components and metastatic lymph nodes in patients with pembrolizumab for first tissue/site agnostic indication. https://www.fda.gov/
resected lung adenosquamous carcinoma. Clin Lung Cancer. 2018;19(4):e421– Drugs/InformationOnDrugs/ApprovedDrugs/ucm560040.htm. Accessed June 15,
e430. 2018.
43. Munari E, Zamboni G, Lunardi G, et al. PD-L1 expression comparison 72. US Food & Drug Administration. FDA grants nivolumab accelerated
between primary and relapsed non-small cell lung carcinoma using whole approval for MSI-H or dMMR colorectal cancer. https://www.fda.gov/drugs/
sections and clone SP263. Oncotarget. 2018;9(54):30465–30471. informationondrugs/approveddrugs/ucm569366.htm. Accessed July 15, 2018.
44. Ilie M, Long-Mira E, Bence C, et al. Comparative study of the PD-L1 status 73. Overman MJ, McDermott R, Leach JL, et al. Nivolumab in patients with
between surgically resected specimens and matched biopsies of NSCLC patients metastatic DNA mismatch repair-deficient or microsatellite instability-high
reveal major discordances: a potential issue for anti-PD-L1 therapeutic strategies. colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study.
Ann Oncol. 2016;27(1):147–153. Lancet Oncol. 2017;18(9):1182–1191.
45. Ribas A, Hu-Lieskovan S. What does PD-L1 positive or negative mean? J 74. Kinzler KW, Vogelstein B. Lessons from hereditary colorectal cancer. Cell.
Exp Med. 2016;213(13):2835–2840. 1996;87(2):159–170.

Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al 721
75. Lin EI, Tseng LH, Gocke CD, et al. Mutational profiling of colorectal 101. Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical
cancers with microsatellite instability. Oncotarget. 2015;6(39):42334–42344. response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371(23):2189–
76. Arends M, Frayling I, Happerfield L, Ibrahim M. HNPCC/Lynch syndrome 2199.
module: report of the immunohistochemical analysis of mismatch repair (MMR) 102. Rizvi NA, Hellmann MD, Snyder A, et al. Mutational landscape
protein expression. UK NEQAS ICC ISH Recomm. 2010;8. determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science.
77. Giardiello FM, Allen JI, Axilbund JE, et al. Guidelines on genetic 2015;348(6230):124–128.
evaluation and management of Lynch syndrome: a consensus statement by the 103. Hellmann MD, Ciuleanu TE, Pluzanski A, et al. Nivolumab plus
US Multi-Society Task Force on colorectal cancer. Gastroenterology. 2014;147(2): ipilimumab in lung cancer with a high tumor mutational burden. N Engl J
502–526. Med. 2018;378(22):2093–2104.
78. Lamberti C, Kruse R, Ruelfs C, et al. Microsatellite instability—a useful 104. Hellmann MD, Nathanson T, Rizvi H, et al. Genomic features of response
diagnostic tool to select patients at high risk for hereditary non-polyposis to combination immunotherapy in patients with advanced non-small-cell lung
colorectal cancer: a study in different groups of patients with colorectal cancer. cancer. Cancer Cell. 2018;33(5):843–852.e844.
Gut. 1999;44(6):839–843. 105. Rosenberg JE, Hoffman-Censits J, Powles T, et al. Atezolizumab in patients
79. Liu T, Wahlberg S, Burek E, Lindblom P, Rubio C, Lindblom A. with locally advanced and metastatic urothelial carcinoma who have progressed
Microsatellite instability as a predictor of a mutation in a DNA mismatch repair following treatment with platinum-based chemotherapy: a single-arm, multi-
gene in familial colorectal cancer. Genes Chromosomes Cancer. 2000;27(1):17– centre, phase 2 trial. Lancet. 2016;387(10031):1909–1920.
25. 106. Goodman AM, Kato S, Bazhenova L, et al. Tumor mutational burden as an
80. Marcus VA, Madlensky L, Gryfe R, et al. Immunohistochemistry for independent predictor of response to immunotherapy in diverse cancers. Mol
hMLH1 and hMSH2: a practical test for DNA mismatch repair-deficient tumors. Cancer Ther. 2017;16(11):2598–2608.
Am J Surg Pathol. 1999;23(10):1248–1255. 107. Carbone DP, Reck M, Paz-Ares L, et al. First-line nivolumab in stage IV or
81. Chen W, Swanson BJ, Frankel WL. Molecular genetics of microsatellite- recurrent non-small-cell lung cancer. N Engl J Med. 2017;376(25):2415–2426.
unstable colorectal cancer for pathologists. Diagn Pathol. 2017;12(1):24. 108. Rizvi H, Sanchez-Vega F, La K, et al. Molecular determinants of response
82. Evaluation of Genomic Applications in Practice and Prevention (EGAPP) to anti-programmed cell death (PD)-1 and anti-programmed death-ligand 1 (PD-
Working Group. Recommendations from the EGAPP Working Group: genetic L1) blockade in patients with non-small-cell lung cancer profiled with targeted
testing strategies in newly diagnosed individuals with colorectal cancer aimed at next-generation sequencing. J Clin Oncol. 2018;36(7):633–641.
reducing morbidity and mortality from Lynch syndrome in relatives. Genet Med. 109. Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100 000 human
2009;11(1):35–41. cancer genomes reveals the landscape of tumor mutational burden. Genome
83. Muller A, Giuffre G, Edmonston TB, et al. Challenges and pitfalls in Med. 2017;9(1):34.
HNPCC screening by microsatellite analysis and immunohistochemistry. J Mol 110. Campesato LF, Barroso-Sousa R, Jimenez L, et al. Comprehensive cancer-
Diagn. 2004;6(4):308–315. gene panels can be used to estimate mutational load and predict clinical benefit
84. Ryan E, Sheahan K, Creavin B, Mohan HM, Winter DC. The current value to PD-1 blockade in clinical practice. Oncotarget. 2015;6(33):34221–34227.
of determining the mismatch repair status of colorectal cancer: a rationale for 111. Ramalingam SS, Hellmann MD, Awad MM, et al. Abstract CT078: Tumor
routine testing. Crit Rev Oncol Hematol. 2017;116:38–57. mutational burden (TMB) as a biomarker for clinical benefit from dual immune
85. Markow M, Chen W, Frankel WL. Immunohistochemical pitfalls: common checkpoint blockade with nivolumab (nivo) þ ipilimumab (ipi) in first-line (1L)
mistakes in the evaluation of Lynch syndrome. Surg Pathol Clin. 2017;10(4):977– non-small cell lung cancer (NSCLC): identification of TMB cutoff from
1007. CheckMate 568. Cancer Res. 2018;78(13)(suppl):CT078. Abstract CT078.
86. Wong HL, Christie M, Gately L, et al. Mismatch repair deficiency
112. Friends of Cancer Research. TMB Harmonization Working Group
assessment by immunohistochemistry: for Lynch syndrome screening and
Meeting. https://www.focr.org/blog/engaging-innovation/tmb-harmonization-
beyond. Future Oncol. 2018;14(26):2725–2739.
working-group-meeting. Accessed August 23, 2018.
87. Shia J, Stadler Z, Weiser MR, et al. Immunohistochemical staining for
113. Gandara DR, Paul SM, Kowanetz M, et al. Blood-based tumor mutational
DNA mismatch repair proteins in intestinal tract carcinoma: how reliable are
burden as a predictor of clinical benefit in non-small-cell lung cancer patients
biopsy samples? Am J Surg Pathol. 2011;35(3):447–454.
treated with atezolizumab. Nat Med. 2018;24(9):1441–1448.
88. O’Brien O, Ryan E, Creavin B, et al. Correlation of immunohistochemical
114. Heitzer E, Tomlinson I. Replicative DNA polymerase mutations in cancer.
mismatch repair protein status between colorectal carcinoma endoscopic biopsy
Curr Opin Genet Dev. 2014;24:107–113.
and resection specimens. J Clin Pathol. 2018;71(7):631–636.
115. Ahn SM, Ansari AA, Kim J, et al. The somatic POLE P286R mutation
89. Bao F, Panarelli NC, Rennert H, Sherr DL, Yantiss RK. Neoadjuvant therapy
defines a unique subclass of colorectal cancer featuring hypermutation,
induces loss of MSH6 expression in colorectal carcinoma. Am J Surg Pathol.
representing a potential genomic biomarker for immunotherapy. Oncotarget.
2010;34(12):1798–1804.
2016;7(42):68638–68649.
90. Vilkin A, Halpern M, Morgenstern S, et al. How reliable is immunohis-
tochemical staining for DNA mismatch repair proteins performed after neo- 116. Erson-Omay EZ, Caglayan AO, Schultz N, et al. Somatic POLE mutations
adjuvant chemoradiation? Hum Pathol. 2014;45(10):2029–2036. cause an ultramutated giant cell high-grade glioma subtype with better prognosis.
91. Toon CW, Chou A, DeSilva K, et al. BRAFV600E immunohistochemistry in Neuro Oncol. 2015;17(10):1356–1364.
conjunction with mismatch repair status predicts survival in patients with 117. Pages F, Mlecnik B, Marliot F, et al. International validation of the
colorectal cancer. Mod Pathol. 2014;27(5):644–650. consensus Immunoscore for the classification of colon cancer: a prognostic and
92. Boland CR, Thibodeau SN, Hamilton SR, et al. A National Cancer Institute accuracy study. Lancet. 2018;391(10135):2128–2139.
workshop on microsatellite instability for cancer detection and familial 118. van Gool IC, Eggink FA, Freeman-Mills L, et al. POLE proofreading
predisposition: development of international criteria for the determination of mutations elicit an antitumor immune response in endometrial cancer. Clin
microsatellite instability in colorectal cancer. Cancer Res. 1998;58(22):5248– Cancer Res. 2015;21(14):3347–3355.
5257. 119. Jansen AM, van Wezel T, van den Akker BE, et al. Combined mismatch
93. Xicola RM, Llor X, Pons E, et al. Performance of different microsatellite repair and POLE/POLD1 defects explain unresolved suspected Lynch syndrome
marker panels for detection of mismatch repair-deficient colorectal tumors. J Natl cancers. Eur J Hum Genet. 2016;24(7):1089–1092.
Cancer Inst. 2007;99(3):244–252. 120. Mertz TM, Baranovskiy AG, Wang J, Tahirov TH, Shcherbakova PV.
94. Hegde M, Ferber M, Mao R, Samowitz W, Ganguly A. ACMG technical Nucleotide selectivity defect and mutator phenotype conferred by a colon
standards and guidelines for genetic testing for inherited colorectal cancer (Lynch cancer-associated DNA polymerase delta mutation in human cells. Oncogene.
syndrome, familial adenomatous polyposis, and MYH-associated polyposis). 2017;36(31):4427–4433.
Genet Med. 2014;16(1):101–116. 121. Mertz TM, Sharma S, Chabes A, Shcherbakova PV. Colon cancer-
95. Gan C, Love C, Beshay V, et al. Applicability of next generation associated mutator DNA polymerase delta variant causes expansion of dNTP
sequencing technology in microsatellite instability testing. Genes (Basel). 2015; pools increasing its own infidelity. Proc Natl Acad Sci U S A. 2015;112(19):
6(1):46–59. E2467–E2476.
96. Salipante SJ, Scroggins SM, Hampel HL, Turner EH, Pritchard CC. 122. Bourdais R, Rousseau B, Pujals A, et al. Polymerase proofreading domain
Microsatellite instability detection by next generation sequencing. Clin Chem. mutations: new opportunities for immunotherapy in hypermutated colorectal
2014;60(9):1192–1199. cancer beyond MMR deficiency. Crit Rev Oncol Hematol. 2017;113:242–248.
97. Nowak JA, Yurgelun MB, Bruce JL, et al. Detection of mismatch repair 123. Mouw KW, Goldberg MS, Konstantinopoulos PA, D’Andrea AD. DNA
deficiency and microsatellite instability in colorectal adenocarcinoma by damage and repair biomarkers of immunotherapy response. Cancer Discov.
targeted next-generation sequencing. J Mol Diagn. 2017;19(1):84–91. 2017;7(7):675–693.
98. Kautto EA, Bonneville R, Miya J, et al. Performance evaluation for rapid 124. Gubin MM, Schreiber RD. The odds of immunotherapy success. Science.
detection of pan-cancer microsatellite instability with MANTIS. Oncotarget. 2015;350(6257):158–159.
2017;8(5):7452–7463. 125. Sharma P, Allison JP. The future of immune checkpoint therapy. Science.
99. Vanderwalde A, Spetzler D, Xiao N, Gatalica Z, Marshall J. Microsatellite 2015;348(6230):56–61.
instability status determined by next-generation sequencing and compared with 126. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy.
PD-L1 and tumor mutational burden in 11 348 patients. Cancer Med. 2018;7(3): Science. 2015;348(6230):69–74.
746–756. 127. Robbins PF, Lu YC, El-Gamil M, et al. Mining exomic sequencing data to
100. Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational identify mutated antigens recognized by adoptively transferred tumor-reactive T
processes in human cancer. Nature. 2013;500(7463):415–421. cells. Nat Med. 2013;19(6):747–752.

722 Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al
128. van Rooij N, van Buuren MM, Philips D, et al. Tumor exome analysis 155. Giraldo NA, Nguyen P, Engle EL, et al. Multidimensional, quantitative
reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive assessment of PD-1/PD-L1 expression in patients with Merkel cell carcinoma and
melanoma. J Clin Oncol. 2013;31(32):e439–e442. association with response to pembrolizumab. J Immunother Cancer. 2018;6(1):
129. Linnemann C, van Buuren MM, Bies L, et al. High-throughput epitope 99.
discovery reveals frequent recognition of neo-antigens by CD4þ T cells in human 156. Johnson DB, Bordeaux J, Kim JY, et al. Quantitative spatial profiling of PD-
melanoma. Nat Med. 2015;21(1):81–85. 1/PD-L1 interaction and HLA-DR/IDO-1 predicts improved outcomes of anti-PD-
130. Lu YC, Yao X, Crystal JS, et al. Efficient identification of mutated cancer 1 therapies in metastatic melanoma. Clin Cancer Res. 2018;24(21):5250–5260.
antigens recognized by T cells associated with durable tumor regressions. Clin 157. Hendry S, Salgado R, Gevaert T, et al. Assessing tumor-infiltrating
Cancer Res. 2014;20(13):3401–3410. lymphocytes in solid tumors: a practical review for pathologists and proposal for
131. Fritsch EF, Rajasagi M, Ott PA, Brusic V, Hacohen N, Wu CJ. HLA-binding a standardized method from the International Immuno-Oncology Biomarkers
properties of tumor neoepitopes in humans. Cancer Immunol Res. 2014;2(6): Working Group: part 2: TILs in melanoma, gastrointestinal tract carcinomas, non-
522–529. small cell lung carcinoma and mesothelioma, endometrial and ovarian
132. Van Bergen CA, Rutten CE, Van Der Meijden ED, et al. High-throughput carcinomas, squamous cell carcinoma of the head and neck, genitourinary
characterization of 10 new minor histocompatibility antigens by whole genome carcinomas, and primary brain tumors. Adv Anat Pathol. 2017;24(6):311–335.
association scanning. Cancer Res. 2010;70(22):9073–9083. 158. Hendry S, Salgado R, Gevaert T, et al. Assessing tumor-infiltrating
133. van Buuren MM, Calis JJ, Schumacher TN. High sensitivity of cancer lymphocytes in solid tumors: a practical review for pathologists and proposal for
exome-based CD8 T cell neo-antigen identification. Oncoimmunology. 2014;3: a standardized method from the International Immunooncology Biomarkers
e28836. Working Group: part 1: assessing the host immune response, TILs in invasive
134. Schumacher T, Bunse L, Pusch S, et al. A vaccine targeting mutant IDH1 breast carcinoma and ductal carcinoma in situ, metastatic tumor deposits and
induces antitumour immunity. Nature. 2014;512(7514):324–327. areas for further research. Adv Anat Pathol. 2017;24(5):235–251.
135. Tran E, Robbins PF, Rosenberg SA. ‘‘Final common pathway’’ of human 159. Salgado R, Denkert C, Demaria S, et al. The evaluation of tumor-
cancer immunotherapy: targeting random somatic mutations. Nat Immunol. infiltrating lymphocytes (TILs) in breast cancer: recommendations by an
2017;18(3):255–262. International TILs Working Group 2014. Ann Oncol. 2015;26(2):259–271.
136. Parkhurst MR, Robbins PF, Tran E, et al. Unique neoantigens arise from 160. Klauschen F, Muller KR, Binder A, et al. Scoring of tumor-infiltrating
somatic mutations in patients with gastrointestinal cancers. Cancer Discov. 2019; lymphocytes: from visual estimation to machine learning. Semin Cancer Biol.
9(8):1022–1035. 2018;52(pt 2):151–157.
137. Zacharakis N, Chinnasamy H, Black M, et al. Immune recognition of 161. Taube JM, Anders RA, Young GD, et al. Colocalization of inflammatory
somatic mutations leading to complete durable regression in metastatic breast response with B7-h1 expression in human melanocytic lesions supports an
cancer. Nat Med. 2018;24(6):724–730. adaptive resistance mechanism of immune escape. Sci Transl Med. 2012;4(127):
138. Stevanovic S, Pasetto A, Helman SR, et al. Landscape of immunogenic 127ra137.
tumor antigens in successful immunotherapy of virally induced epithelial cancer. 162. Taube JM, Young GD, McMiller TL, et al. Differential expression of
Science. 2017;356(6334):200–205. immune-regulatory genes associated with PD-L1 display in melanoma: implica-
139. Mahmoud SM, Paish EC, Powe DG, et al. Tumor-infiltrating CD8þ tions for PD-1 pathway blockade. Clin Cancer Res. 2015;21(17):3969–3976.
lymphocytes predict clinical outcome in breast cancer. J Clin Oncol. 2011; 163. Lipson EJ, Vincent JG, Loyo M, et al. PD-L1 expression in the Merkel cell
29(15):1949–1955. carcinoma microenvironment: association with inflammation, Merkel cell
140. Mlecnik B, Tosolini M, Kirilovsky A, et al. Histopathologic-based polyomavirus and overall survival. Cancer Immunol Res. 2013;1(1):54–63.
prognostic factors of colorectal cancers are associated with the state of the local 164. Velcheti V, Schalper KA, Carvajal DE, et al. Programmed death ligand-1
immune reaction. J Clin Oncol. 2011;29(6):610–618. expression in non-small cell lung cancer. Lab Invest. 2014;94(1):107–116.
141. Rakaee M, Kilvaer TK, Dalen SM, et al. Evaluation of tumor-infiltrating 165. Cimino-Mathews A, Thompson E, Taube JM, et al. PD-L1 (B7-H1)
lymphocytes using routine H&E slides predicts patient survival in resected non- expression and the immune tumor microenvironment in primary and metastatic
small cell lung cancer. Hum Pathol. 2018;79:188–198. breast carcinomas. Hum Pathol. 2016;47(1):52–63.
142. Smyrk TC, Watson P, Kaul K, Lynch HT. Tumor-infiltrating lymphocytes are 166. Schalper KA, Velcheti V, Carvajal D, et al. In situ tumor PD-L1 mRNA
a marker for microsatellite instability in colorectal carcinoma. Cancer. 2001; expression is associated with increased TILs and better outcome in breast
91(12):2417–2422. carcinomas. Clin Cancer Res. 2014;20(10):2773–2782.
143. Chen PL, Roh W, Reuben A, et al. Analysis of immune signatures in 167. Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations associated with
longitudinal tumor samples yields insight into biomarkers of response and acquired resistance to PD-1 blockade in melanoma. N Engl J Med. 2016;375(9):
mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 2016; 819–829.
6(8):827–837. 168. Ribas A, Robert C, Hodi FS, et al. Association of response to programmed
144. Wickenhauser C, Bethmann D, Feng Z, et al. Multispectral fluorescence death receptor 1 (PD-1) blockade with pembrolizumab (MK-3475) with an
imaging allows for distinctive topographic assessment and subclassification of interferon-inflammatory immune gene signature. J Clin Oncol. 2015;
tumor-infiltrating and surrounding immune cells. Methods Mol Biol. 2019;1913: 33(15)(suppl):3001.
13–31. 169. Fehrenbacher L, Spira A, Ballinger M, et al. Atezolizumab versus
145. Hofman P, Badoual C, Henderson F, et al. Multiplexed immunohisto- docetaxel for patients with previously treated non-small-cell lung cancer
chemistry for molecular and immune profiling in lung cancer—just about ready (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial.
for prime-time? Cancers (Basel). 2019;11(3). Lancet. 2016;387(10030):1837–1846.
146. Parra ER, Francisco-Cruz A, Wistuba II. State-of-the-art of profiling 170. Shin DS, Zaretsky JM, Escuin-Ordinas H, et al. Primary resistance to PD-1
immune contexture in the era of multiplexed staining and digital analysis to study blockade mediated by JAK1/2 mutations. Cancer Discov. 2017;7(2):188–201.
paraffin tumor tissues. Cancers (Basel). 2019;11(2). 171. Gettinger S, Choi J, Hastings K, et al. Impaired HLA class I antigen
147. Remark R, Merghoub T, Grabe N, et al. In-depth tissue profiling using processing and presentation as a mechanism of acquired resistance to immune
multiplexed immunohistochemical consecutive staining on single slide. Sci checkpoint inhibitors in lung cancer. Cancer Discov. 2017;7(12):1420–1435.
Immunol. 2016;1(1):aaf6925. 172. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic beta-catenin
148. Day WA, Lefever MR, Ochs RL, et al. Covalently deposited dyes: a new signalling prevents anti-tumour immunity. Nature. 2015;523(7559):231–235.
chromogen paradigm that facilitates analysis of multiple biomarkers in situ. Lab 173. Peng W, Chen JQ, Liu C, et al. Loss of PTEN promotes resistance to T cell-
Invest. 2017;97(1):104–113. mediated immunotherapy. Cancer Discov. 2016;6(2):202–216.
149. Gerdes MJ, Sevinsky CJ, Sood A, et al. Highly multiplexed single-cell 174. Gainor JF, Shaw AT, Sequist LV, et al. EGFR mutations and ALK
analysis of formalin-fixed, paraffin-embedded cancer tissue. Proc Natl Acad Sci U rearrangements are associated with low response rates to PD-1 pathway blockade
S A. 2013;110(29):11982–11987. in non-small cell lung cancer: a retrospective analysis. Clin Cancer Res. 2016;
150. Buchwalow I, Samoilova V, Boecker W, Tiemann M. Multiple 22(18):4585–4593.
immunolabeling with antibodies from the same host species in combination 175. Hugo W, Zaretsky JM, Sun L, et al. Genomic and transcriptomic features of
with tyramide signal amplification. Acta Histochem. 2018;120(5):405–411. response to anti-PD-1 therapy in metastatic melanoma. Cell. 2016;165(1):35–44.
151. Zhang W, Hubbard A, Jones T, et al. Fully automated 5-plex fluorescent 176. Blank CU, Haanen JB, Ribas A, Schumacher TN. The ‘‘cancer immuno-
immunohistochemistry with tyramide signal amplification and same species gram.’’ Science. 2016;352(6286):658–660.
antibodies. Lab Invest. 2017;97(7):873–885. 177. Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences
152. Hagemann AR, Cadungog M, Hagemann IS, et al. Tissue-based immune efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;
monitoring I: tumor core needle biopsies allow in-depth interrogation of the 359(6371):91–97.
tumor microenvironment. Cancer Biol Ther. 2011;12(4):357–366. 178. Matson V, Fessler J, Bao R, et al. The commensal microbiome is associated
153. Mezheyeuski A, Bergsland CH, Backman M, et al. Multispectral imaging with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;
for quantitative and compartment-specific immune infiltrates reveals distinct 359(6371):104–108.
immune profiles that classify lung cancer patients. J Pathol. 2018;244(4):421– 179. Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome modulates
431. response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;
154. Siska PJ, Johnpulle RAN, Zhou A, et al. Deep exploration of the immune 359(6371):97–103.
infiltrate and outcome prediction in testicular cancer by quantitative multiplexed 180. Parsa AT, Waldron JS, Panner A, et al. Loss of tumor suppressor PTEN
immunohistochemistry and gene expression profiling. Oncoimmunology. 2017; function increases B7-H1 expression and immunoresistance in glioma. Nat Med.
6(4):e1305535. 2007;13(1):84–88.

Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al 723
181. Marzec M, Zhang Q, Goradia A, et al. Oncogenic kinase NPM/ALK 185. Goodman AM, Piccioni D, Kato S, et al. Prevalence of PDL1 amplification
induces through STAT3 expression of immunosuppressive protein CD274 (PD-L1, and preliminary response to immune checkpoint blockade in solid tumors. JAMA
B7-H1). Proc Natl Acad Sci U S A. 2008;105(52):20852–20857. Oncol. 2018;4(9):1237–1244.
182. Ota K, Azuma K, Kawahara A, et al. Induction of PD-L1 expression by the 186. Kataoka K, Shiraishi Y, Takeda Y, et al. Aberrant PD-L1 expression through
EML4-ALK oncoprotein and downstream signaling pathways in non-small cell 3 0 -UTR disruption in multiple cancers. Nature. 2016;534(7607):402–406.
lung cancer. Clin Cancer Res. 2015;21(17):4014–4021. 187. Coelho MA, de Carne Trecesson S, Rana S, et al. Oncogenic RAS signaling
183. Roemer MG, Advani RH, Ligon AH, et al. PD-L1 and PD-L2 genetic promotes tumor immunoresistance by stabilizing PD-L1 mRNA. Immunity. 2017;
alterations define classical Hodgkin lymphoma and predict outcome. J Clin 47(6):1083–1099.e1086.
Oncol. 2016;34(23):2690–2697. 188. Peters S, Creelan B, Hellmann MD, et al. Impact of tumor mutation burden
184. Chapuy B, Roemer MG, Stewart C, et al. Targetable genetic features of on the efficacy of first-line nivolumab in stage iv or recurrent non-small cell lung
primary testicular and primary central nervous system lymphomas. Blood. 2016; cancer: an exploratory analysis of CheckMate 026. Cancer Res. 2017;
127(7):869–881. 77(13)(suppl):CT082. Abstract CT082.

724 Arch Pathol Lab Med—Vol 144, June 2020 Cancer Immunotherapy Biomarker Testing Landscape—Walk et al

You might also like