The development of immunotherapy is an important breakthrough for the treatment of cancer, with antitumor efficacy observed in a wide variety of tumors. To optimize immunotherapy use, approaches must be developed to identify which patients are likely to achieve benefit. To minimize therapeutic toxicities and costs, understanding the ideal choice and sequencing of the numerous immuno-oncology agents available for individual patients is thus critical, but fraught with challenges. The immune tumor microenvironment (TME) is a unique aspect of the response to immuno-oncology agents and measurement of single biomarkers does not adequately capture these complex interactions. Therefore, multiple potential biomarkers are likely needed. Current candidates in this area include PD-L1 expression, CD8+ tumor-infiltrating lymphocytes, tumor mutation load and neoantigen burden, immune-related gene signatures, and multiplex IHC assays that examine the pharmacodynamic and spatial interactions of the TME. The most fruitful investigations are likely to use several techniques to predict response and interrogate mechanisms of resistance. Immuno-oncology biomarker research must employ validated assays to ask focused research questions utilizing clinically annotated tissue collections and biomarker-focused clinical trial designs to investigate specific endpoints. Real-time input from patients and their advocates into biomarker discovery is necessary to ensure that the investigations pursued will improve both clinical outcomes and quality of life. We herein provide a framework of recommendations to guide the search for immuno-oncology biomarkers of value. Clin Cancer Res; 23(17); 4970–9. ©2017 AACR.

See all articles in this CCR Focus section, “Clinical Trial Design Considerations in the Immuno-oncology Era.”

Checkpoint inhibitors are emerging as among the most effective anticancer agents in our armamentarium. These agents, which inhibit the immune checkpoint proteins programmed death cell protein-1 (PD-1) or CTL associated antigen 4 (CTLA-4) are able to produce durable long-term remissions and are active in a broad range of tumor types, although not all patients respond (1–7). While antitumor activity is seen across a wide range of histologies, the response rate varies by tumor type, with responses to single-agent PD-1 inhibitor therapy in the 50%–60% range in Merkel cell carcinoma (8) and relapsed Hodgkin lymphoma (9), in the 30%–40% range for melanoma and non–small cell lung cancers (10, 11), and in the 10%–20% range for multiple solid tumors in early-phase trials (12, 13). Combinatorial strategies may improve response rates but also increase toxicity and should be approached with caution given the significant immune-mediated toxicities than can develop (11). Thus, biomarkers that predict response, resistance, or toxicity are of paramount importance to effectively develop these agents. This article will discuss the challenges of immune-related biomarkers that relate to the unique science of the dynamic immune microenvironment, the lack of available tissue collections with attention to specimen quality, strategies for incorporation of biomarker endpoints into clinical trials, and the critical importance of the patient's perspective, accompanied by recommendations to provide a framework for further investigation and discussion.

The immune system is in constant flux, with targets that vary based on specific time points and locations within tumors, the presence of "microniches" (created by alterations in perfusion or oxygenation) and multiple different populations of immune-infiltrating cells (refs. 14, 15; Fig. 1). Thus, unlike the evaluation of certain classes of targeted therapies that inhibit specific signaling targets, identifying critical immune interactions to measure at given points in this heterogenous environment may be difficult, rendering biomarker development uniquely challenging. In addition, immuno-oncology biomarkers collected at various time points in a disease course will serve different purposes, for instance, prediction of response versus monitoring for relapse; thus, different clinical scenarios confer different implications for biomarker development.

Figure 1.

Putative immuno-oncology biomarkers in the TME. 1. PD-L1 expression: PD-L1 expression in the tumor microenvironment may indicate increased responsiveness to blockade of the PD-1/PD-L1 checkpoint. 2. Tumor-infiltrating lymphocytes (TILs): the presence of tumor-infiltrating lymphocytes may be indicative of a preexisting antitumor immune response that can be reinvigorated by immunotherapy. 3. Mutational load and neoantigens: increased tumor mutational load and putative neoantigens may be a marker for increased immunogenicity of a tumor. 4. Immunosuppressive cell types: immunosuppressive cells, such as immature dendritic cells, myeloid-derived suppressor cells (MDSC), tumor-associated macrophages, and T-regulatory cells are recruited to or generated in the tumor microenvironment. The presence of these cell types may indicate resistance or sensitivity to specific types of therapy. 5. Macrophage and DC polarization: macrophages can be proinflammatory (M1) or anti-inflammatory (M2). M2 macrophages and MDSCs inhibit T-cell responses through a variety of mechanisms and their presence may indicate resistance to certain types of immunotherapy. Dendritic cells (DC) can also be polarized from immature into primed immunosuppressive/tolerogenic regulatory DCs, which limit activity of effector T cells and support tumor growth and progression. 6. Immunosuppressive molecules: the presence of other modes of immune suppression such as PD-1, PD-L1, CTLA-4, IDO, Tim-3 and others may indicate nonoverlapping mechanisms of immune resistance which may predict sensitivity or resistance to immunotherapy. 7. Cytokine signatures: the presence of immunostimulatory or immunoinhibitory cytokines in the tumor microenvironment may predict sensitivity or resistance. Multiple methods exist to interrogate the tumor microenvironment including: IHC/IF, whole exome sequencing (WES) transcriptome analysis, proteomics, flow cytometry and others.

Figure 1.

Putative immuno-oncology biomarkers in the TME. 1. PD-L1 expression: PD-L1 expression in the tumor microenvironment may indicate increased responsiveness to blockade of the PD-1/PD-L1 checkpoint. 2. Tumor-infiltrating lymphocytes (TILs): the presence of tumor-infiltrating lymphocytes may be indicative of a preexisting antitumor immune response that can be reinvigorated by immunotherapy. 3. Mutational load and neoantigens: increased tumor mutational load and putative neoantigens may be a marker for increased immunogenicity of a tumor. 4. Immunosuppressive cell types: immunosuppressive cells, such as immature dendritic cells, myeloid-derived suppressor cells (MDSC), tumor-associated macrophages, and T-regulatory cells are recruited to or generated in the tumor microenvironment. The presence of these cell types may indicate resistance or sensitivity to specific types of therapy. 5. Macrophage and DC polarization: macrophages can be proinflammatory (M1) or anti-inflammatory (M2). M2 macrophages and MDSCs inhibit T-cell responses through a variety of mechanisms and their presence may indicate resistance to certain types of immunotherapy. Dendritic cells (DC) can also be polarized from immature into primed immunosuppressive/tolerogenic regulatory DCs, which limit activity of effector T cells and support tumor growth and progression. 6. Immunosuppressive molecules: the presence of other modes of immune suppression such as PD-1, PD-L1, CTLA-4, IDO, Tim-3 and others may indicate nonoverlapping mechanisms of immune resistance which may predict sensitivity or resistance to immunotherapy. 7. Cytokine signatures: the presence of immunostimulatory or immunoinhibitory cytokines in the tumor microenvironment may predict sensitivity or resistance. Multiple methods exist to interrogate the tumor microenvironment including: IHC/IF, whole exome sequencing (WES) transcriptome analysis, proteomics, flow cytometry and others.

Close modal

Candidate biomarkers include markers of a preexisting antitumor immune infiltrate that is observed in certain developing tumors, but also may be seen in response to both immune and cytotoxic therapies (14, 15). Response to immunotherapy has been linked to an “inflamed” tumor microenvironment (TME), hallmarks of which include expression of IFN-inducible immunosuppressive molecules such as PD-L1 and indoleamine 2,3-dioxygenase (IDO; Fig 1, items 1 and 6), IFNγ production, M1 macrophages, and a robust T-cell infiltrate (Fig. 1, item 2) and may have fewer immunosuppressive cells such as M2 macrophages and myeloid-derived suppressor cells (Fig. 1, items 4, 5; refs. 16, 17). Gene signatures associated with T-cell–inflamed tumors have also predicted response (Fig. 1, item 7; ref. 18). The presence of tumor-infiltrating lymphocytes (TIL) in the TME is mechanistically a logical biomarker for PD-1/PD-L1 inhibitor response, as without these cells, no immune response will occur. Examining the TME from patients treated with CTLA-4 or PD-1/PD-L1 checkpoint blockade demonstrates an association between TIL counts and response both before and after treatment (19, 20). However, these findings are not universal, and the dynamic and individual nature of the response renders these investigations difficult. TILs are known to be heterogenous and ideal methods of measurement can be a topic of debate, with hematoxylin/eosin (H&E) stain being a traditional method but with multiplex and molecular methods holding promise (21). Further work is needed to define optimal cut-off points and other scoring metrics to determine the usefulness of TILs as a response biomarker.

Utilizing the tumor microenvironment to guide therapy

With several trials targeting the PD-1/PD-L1 checkpoint demonstrating that TME PD-L1 expression is positively associated with response (3, 11, 22), PD-L1 testing has begun to be incorporated into clinical algorithms. PD-L1 testing has been developed as a companion diagnostic per FDA guidelines for the treatment of non–small cell lung cancer (23). However, implementation of widespread testing is limited by the magnitude of benefit, which varies considerably between trials and tumor histologies. It is important to note that the shortcomings of PD-L1 testing in current clinical practice are likely related to the variability of the assays and antibodies used to detect PD-L1, the thresholds used to define positivity, and the TME cell types that express PD-L1 (Fig. 1, items 1, 6). Thus, while it seems obvious that PD-L1 on either tumor or immune cells (24, 25) must be present for immune checkpoint therapy to be effective and tumors that are truly negative for PD-L1 should be resistant to therapy, the result from a PD-L1 test may be inaccurate and an apparently negative tumor may still respond (3, 22, 26, 27). Ultimately, while PD-L1 expression may enrich for the potential of obtaining treatment response, it alone cannot adequately summarize the complexity of the tumor–immune system interactions and consistently predict patient benefit from immunotherapy. In tumors with constitutive PD-L1 (B7-H1) expression, the predictive value of PD-L1 may be improved by adding additional parameters such as infiltrating CD8+ T cells or an IFNγ gene signature.

In dissecting the tumor microenvironment to understand which tumors may be most responsive to immunotherapy, the “Tumor-Immune Microenvironment (TIME)” classification system, developed by Zhang and Chen is helpful (28). This system builds upon prior classifications of tumors as T-cell inflamed, infiltrated with T lymphocytes, and an IFN signature that may be primed for response to immunotherapy, versus noninflamed tumors that lack robust T-cell infiltration and are more resistant to immune approaches (17, 29, 30). TIME recognizes 4 separate phenotypes within the TME, T1 (B7-H1, TIL), T2 (B7-H1+, TIL+), T3 (B7-H1, TIL+), and T4 (B7-H1+, TIL), which segregate tumors that are responsive from those that are resistant to checkpoint blockade (see Fig. 2; modeled after the figure in ref. 28 for an illustration of these phenotypes). T2 tumors, which typically contain TILs and other immune cells, but are resistant to cell killing by TILs due to B7-H1 expression, are thought to account for most anti-PD-1 responses. The T2 tumors that are resistant to anti-PD-1 therapy must have additional dominant immune pathways that are not fully understood and the source of active research. T1 and T4 tumors account for most tumors resistant to anti-PD-1 therapy, as they both lack TILs, but T4 differs from T1 in that it has intrinsic B7-H1 expression, likely due to the activation of oncogenic pathways (31). T3 tumors have TILs, but lack B7-H1 expression, likely owing to the absence of IFNγ production by T effector cells; similar to T1 tumors, these are likely to be resistant to anti-PD-1 therapy and may require agents with other mechanisms of action to affect the TME.

Figure 2.

Subtypes of TIME classification. This figure, modeled after Zhang and Chen's (28), illustrates the four tumor subtypes of TIME; T1–T4.

Figure 2.

Subtypes of TIME classification. This figure, modeled after Zhang and Chen's (28), illustrates the four tumor subtypes of TIME; T1–T4.

Close modal

Impact of genomic factors on response to immunotherapy

Tumor-specific genomic factors also play a role in response to immunotherapy (Fig. 1, item 3). Mutational burden is the nonsynonymous somatic mutational load of a tumor measured by DNA sequencing and serves as a surrogate for the number of mutated proteins within a tumor. This is thought to reflect the number of “non-self” antigens that the tumor contains, which is itself a surrogate for tumor antigenicity. The increased mutational burden of microsatellite unstable tumors is believed to account for their sensitivity to checkpoint inhibitors (32). In addition, tumors that harbor defects in enzymes such as DNA polymerase epsilon (POLE) may have an “ultramutator phenotype” that is especially vulnerable to checkpoint inhibition (33), a phenomenon similarly identified through investigation of exceptional responders. Furthermore, mutational burden has been associated with improved response to these agents in NSCLC (6) and melanoma (34–36) and recent data suggest that accounting for both the magnitude of mutations and their expression across tumor cell clones is more prognostic than mutational burden alone (37). The widespread expression of neoantigens across the heterogeneous tumor means that a T-cell response to that antigen is more likely to develop, and that a larger proportion of tumor cells are susceptible to attack by T cells that recognize that antigen. Complicating issues with the applicability of this approach include ready availability of adequate tissue for sequencing, variability introduced by tumor location, whether the tumor is primary or metastatic, and the impact of intervening treatments on sequencing results.

High mutation burden is not the only mechanism which induces local immune activation. Many human cancers are associated with viral infections such as Epstein–Barr virus (EBV) and human papillomavirus (HPV). As viral proteins can be strongly antigenic and lead to a local immune response requiring induction of immune checkpoints for tumor survival, it stands to reason that viral-associated cancers may be more immunogenic and more responsive to immunotherapy. Besides antigenicity, viral proteins can also influence the interaction between tumor and immune system through several other mechanisms. For example, patients with latent cytomegalovirus may have increased antitumor cytotoxic activity of natural killer cells (38). Likewise, EBV may drive the expression of PD-L1 (independent of 9p24.1 amplification) in Hodgkin lymphoma and other EBV+ cancers (39, 40). Interestingly, Hodgkin lymphoma is one of the most responsive histologies to checkpoint blockade and expresses high levels of PD-L1 (41). In head and neck cancers, HPV+ tumors may have less PD-L1 expression but a higher proportion of T-cell infiltration (42). Thus, the presence of viral proteins could serve as an important biomarker for immunotherapy but the exact influence may be tumor- and virus-specific. To date, there is little clinical data showing a clear relationship between immunotherapy response and the presence of a viral-associated cancer. In a recent clinical trial testing PD-1 inhibition for Merkel cell carcinoma, PD-L1 expression was more frequent in Merkel cell polyoma virus–positive tumors (71% vs. 25%) and virus-positive tumors had a higher response rate (65% vs. 44%), although the small patient number limits interpretation (8). More research is required to determine whether virus-associated cancers are more responsive to immunotherapy and whether viral proteins can be used as biomarkers for this therapy. It is possible that viral proteins may be useful surrogates for other biomarkers (e.g., PD-L1, TIL density) that reflect the underlying mechanism whereby viral proteins influence the tumor–immune interaction.

Developing research focuses on novel biomarkers

Biomarkers of toxicity have been notoriously difficult to identify; however, recent studies suggest that the immune microbiome plays a role in the development of toxicity (43–45). The microbiome may also be related to response to immune therapy, as seen in a recent report of a larger cohort of 228 patients with metastatic melanoma, nearly half of whom had been treated with anti-PD-1 therapy. In this study, patients who responded to therapy harbored gut microbiota with greater diversity, particularly greater amounts of clostridia bacterium, and tumors from these responding patients showed significantly increased immune infiltrates. As well, a study of fecal samples from 105 patients treated with immune checkpoint blockade indicated that patients with more varied bacteria in their digestive tracts had longer median progression free survival, with the presence or absence of specific bacterial types also influencing outcomes (46). In contrast, among saliva samples analyzed from patients with squamous carcinoma of the head and neck, no significant associations were detected among bacterial diversity with best overall response, tumor PD-L1 expression, or HPV16 status (47). These findings underscore the importance of taking both the tumor and host factors into consideration in the search for biomarkers (48), with further studies needed to determine the relevance of the immune microbiome in predicting toxicity from and response to immunotherapy.

As immuno-oncology research continues to develop, many novel biomarkers are being explored. T-cell clonal diversity, as measured by T-cell receptor deep sequencing, is a measure of the breadth of the T-cell response that has been linked with response to checkpoint inhibition (20, 49). Gene expression profiles, analyzed through proteomic nanostring or RNA-Sequencing approaches, may also be related to the immune response, as has been demonstrated with the use of IFN signatures in melanoma patients treated with pembrolizumab. These signatures correlated with improved clinical outcomes and thus may be ultimately useful for improved patient selection for therapy (18). Exploration of patient factors such as age, sex, and obesity, which may influence immune function, may identify additional biomarkers. As the sophistication and sensitivity of immune assays improve, exploration of peripheral blood immunophenotype and systemic cytokines may reveal prognostic and predictive signatures, and is particularly important given the advantages of blood based biomarkers over tumor biopsies that are invasive, sometimes difficult to procure, and limited by tumor heterogeneity. Proposed approaches include peripheral blood flow cytometry to analyze the immunome, examining PD-1/PD-L1 exosomal material in plasma as a biomarker of immune evasion, and measuring circulating antibodies with specificity for tumor antigens as serologic markers of immune response (17, 50).

While a detailed discussion is outside the scope of this article, a brief mention of imaging biomarkers in development is important as a novel and developing field. De Vries and colleagues have utilized several imaging techniques to study early treatment-related immune processes after myeloid dendritic cell vaccine injection. Dendritic cells have been tracked with both scintography (indium-111 labeled) and MRI (superparamagnetic iron oxide formulations) to study vaccine delivery and intranodal migration patterns of dendritic cells (51). PET scans are also utilized that employ radiolabeled 3′-fluoro-3′-deoxy-thymidine (18F-FLT), a thymidine analogue that incorporates into the DNA of proliferating cells at different post vaccination time points after vaccination to visualize immune responses in the lymph nodes. Selective 18F-FLT uptake may indicate a response parameter, with lack of uptake predicting a lack of response (see “Imaging Inflammation” in ref. 50). Further study of these and other immune-PET techniques that are being developed preclinically (52) are of great interest.

To summarize, it is important to note that no single accurate immuno-oncology biomarker is available to select patients for therapy (Table 1). Perhaps, a composite approach of multiple biomarkers will ultimately be implemented. While combining current developing biomarkers of immune monitoring may improve upon the current landscape, given the fluid nature of the immune milieu, reliable biomarkers are inherently difficult to identify. Further work is needed in these areas and is ongoing.

Table 1.

Clinical significance and challenges of biomarkers for immunotherapy in development

Biomarker/techniqueClinical significanceChallenges
All Predict response, resistance, or toxicity to immune therapies Dynamic immune microenvironment, heterogeneity due to biopsy location, type, and primary versus metastatic lesion, impact of intervening therapies 
PD-L1 (3, 10, 25) IHC approach to measure PD-L1 expression on tumor and immune cells Variability in assays, antibodies, and TME cell types detected 
CD8+ T cells (11, 19, 20) PD-1/PD-L1 expression on these cells predicts response to PD-1 blockade Optimal cut-off points, scoring metrics, and agreement on magnitude of change required for meaningful prediction of response 
Tumor mutation load/WES (6, 33–37) High mutation load resulting from various factors (environmental insults, DNA repair defects) correlated with vulnerability to checkpoint inhibitor therapy in exceptional responders Availability of adequate tissue for sequencing, WES costly with slower turnaround time than many clinical assays 
Neoantigen burden (6, 35–58) Predict clinical benefit to ipilimumab and PD-1 blockade in lung cancer and melanoma Dependent on WES data, with similar obstacles of cost and time 
T-cell clonal diversity (20, 49) T-cell receptor deep sequencing to measure breadth of immune response Availability of adequate tissue for sequencing, preidentification of recognized antigens may be required for further investigations 
Multiplex IHC (55, 59, 60) Immunofluorescent detection of multiple immune cell and tumor phenotypes simultaneously as well as evaluation of spatial relationships within TME Rigorous preassay calibration required; immuno-oncology agents may not affect all markers in the multiplex assay equally and primary markers must be selected 
Microbiome modulation (44–48) Components of the gut microbiota may facilitate the antitumor efficacy of immune checkpoint blockade Inter-patient variability within the microbiome contributes to substantial heterogeneity in the mounting of immune responses 
Gene expression profiling (18–20) IFNγ-induced signatures especially may predict clinical benefit to checkpoint inhibition Sizable tissue collections necessary to validate testing and training sets 
Biomarker/techniqueClinical significanceChallenges
All Predict response, resistance, or toxicity to immune therapies Dynamic immune microenvironment, heterogeneity due to biopsy location, type, and primary versus metastatic lesion, impact of intervening therapies 
PD-L1 (3, 10, 25) IHC approach to measure PD-L1 expression on tumor and immune cells Variability in assays, antibodies, and TME cell types detected 
CD8+ T cells (11, 19, 20) PD-1/PD-L1 expression on these cells predicts response to PD-1 blockade Optimal cut-off points, scoring metrics, and agreement on magnitude of change required for meaningful prediction of response 
Tumor mutation load/WES (6, 33–37) High mutation load resulting from various factors (environmental insults, DNA repair defects) correlated with vulnerability to checkpoint inhibitor therapy in exceptional responders Availability of adequate tissue for sequencing, WES costly with slower turnaround time than many clinical assays 
Neoantigen burden (6, 35–58) Predict clinical benefit to ipilimumab and PD-1 blockade in lung cancer and melanoma Dependent on WES data, with similar obstacles of cost and time 
T-cell clonal diversity (20, 49) T-cell receptor deep sequencing to measure breadth of immune response Availability of adequate tissue for sequencing, preidentification of recognized antigens may be required for further investigations 
Multiplex IHC (55, 59, 60) Immunofluorescent detection of multiple immune cell and tumor phenotypes simultaneously as well as evaluation of spatial relationships within TME Rigorous preassay calibration required; immuno-oncology agents may not affect all markers in the multiplex assay equally and primary markers must be selected 
Microbiome modulation (44–48) Components of the gut microbiota may facilitate the antitumor efficacy of immune checkpoint blockade Inter-patient variability within the microbiome contributes to substantial heterogeneity in the mounting of immune responses 
Gene expression profiling (18–20) IFNγ-induced signatures especially may predict clinical benefit to checkpoint inhibition Sizable tissue collections necessary to validate testing and training sets 

While immuno-oncology biomarker discovery faces unique challenges, many parallels can be drawn between these efforts and the study of omics-based biomarkers in the setting of clinical trials (53, 54). Conditions of specimen collection, processing and storage must be identified and specimen quality, quantity, and composition rigorously evaluated. Feasibility studies are often necessary prior to incorporating biomarkers into clinical trials, or in settings where the tissue analyzed is irreplaceable. Standard operating procedures and quality assurance protocols must be developed that consider accuracy, precision, sensitivity, and specificity, and turnaround time of assessments must be within acceptable timeframes (53–55).

The above considerations are important for rigorous evaluation of biomarkers that predict both response and primary resistance, as described above, as well as acquired (secondary) resistance. In examining biopsies of tumor tissue, first and foremost it is important to recall that the timing and location of such biopsies are critical in analyzing results and minimizing variability (17). In addition to an on-treatment biopsy to assess parameters such as TIL infiltration, a biopsy may be timed before response is expected to measure viable tumor and changes in lymphocyte infiltration, with an expected appropriate timepoint of 2–3 weeks posttreatment (17). In addition, biopsies repeated at the time of disease progression are highly informative, but often not completed. The reasons for this are multifactorial: patients whose tumors are progressing may be too distressed about the recurrence to consider another procedure and both physicians and patients may feel pressure to start a new therapy. In addition, many early-phase immunotherapy studies have not requested or mandated these biopsies, a potential shortcoming. In part, due to the restrictive nature of clinical trial eligibility (56), many patients who must receive immunotherapy as standard of care may have limited opportunities to participate in research directed at studies of resistance, as such studies may be infrequent or under-resourced. A creative solution to this has been the efforts of patient advocacy groups to begin international tissue bank consortia, for instance in melanoma (57). Finally, immuno-oncology agents induce unique changes in the TME that may lower the diagnostic yield and influence tissue quality of biopsies.

An example of the power of this approach is exemplified by work from the laboratories of Ribas and Lo with their seminal discovery of JAK-2 mutations in melanoma patients at the time of progression on pembrolizumab therapy (58). In this study, the authors performed whole-exome sequencing using the NimbleGen SeqCap EZ Human Exome Library followed by targeted gene expression of genes revealed by whole exome sequencing, using the nCountersystem (NanoString Technologies) to analyze biopsies from melanoma patients upon initiation of pembrolizumab therapy and at the time of development of resistance. Established human melanoma cell lines from patients were also used to analyze recognition by T-cell receptor transgenic T cells with the use of in vitro coculture assays that detect antigen-induced release of IFNγ. Resistance-associated loss-of-function mutations in the genes encoding IFN receptor–associated Janus kinase 1 or 2 (JAK1 or JAK2), concurrent with deletion of the wild-type allele, resulted in a lack of response to IFNγ and insensitivity to its antiproliferative effect on tumor cells. A truncating mutation in the gene encoding the antigen-presenting protein beta-2-microglobulin (B2M) was also identified in a third patient and led to loss of surface expression of MHC class I. This elegant study suggests that tumors develop resistance to immune checkpoint blockade by unanticipated mechanisms and further emphasizes the importance of tumor biopsies at the time of progression to truly understand in vivo biology.

Newer approaches, including image-based in vivo detection, may allow further characterization of the TME and more comprehensive examination of pharmacodynamic effects (ref. 59; Table 1). Multiplexed IHC assays that utilize novel microscopy and image analysis techniques can interrogate multiple antigens and their interactions within the TME. These analyses allow examination of the atypical dose–response relationships observed in immuno-oncology, where standard pharmacokinetic and pharmacodynamic parameters may not apply (60, 61). Multiplex IHC techniques enable quantitative assessment of biomarkers in multiple cell types and their spatial relationships within tissue, facilitating the examination of multiple potential biomarkers simultaneously (e.g., IDO, LAG3, Tim-3, PD-L1, CTLA-4). The information provided by these techniques may reveal insights into the cell–cell interactions that are important for response to immunomodulating agents and provide novel biomarkers for study in clinical trials. Particular attention to reagent validation and assay calibration is necessary to carry out these techniques, and the impact of tissue quantity and quality (i.e., liquid or small, fine-needle aspirate biopsies vs. core biopsies) cannot be underestimated (55, 62).

In undertaking the above analyses, the availability of collections of clinically annotated tissue for biomarker studies is a critical need, exemplified by the recommendations of the Cancer Moonshot panel (63). By sharing tissue collections from clinical trials and other sources, computational evaluation of multiple biomarkers using high-dimensional correlative data to tease out patterns predictive of response or resistance may be of value (64). The cost and complexity of tissue acquisition and storage and clinical annotation of these collections with evolving clinical outcomes are major obstacles, and the lack of incentive to share resources is a significant limitation. The assembly of cancer immunotherapy networks, and education of patients and providers about the critical importance of tissue collection and analysis, provide important opportunities for advancement.

An important way to optimize the study of biomarkers for immunotherapy, given the challenges set forth above and the utilization of valuable tissues with finite availability, is the careful selection of biomarker endpoints to explore when designing clinical protocols. Prognostic biomarkers, which may be of use in defining a high-risk population in particular need of treatment, and predictive biomarkers that indicate whether a patient will benefit from a particular treatment (53, 65–68), are needed. Choice of trial design must be carefully considered as there are a number of designs for incorporating biomarkers into phase III trials, depending on the predictive power of the biomarker (66, 67, 69–71). The simplest designs are the “enrichment” and “stratified” designs. If it is reasonably clear that treatment benefit is limited to the biomarker positive subgroup, the enrichment design is the most efficient and ethical. If there is doubt about whether the treatment benefit is limited to the biomarker subgroup, or there is doubt that the biomarker is accurate for that group, then the biomarker-stratified design is preferred, although it may increase the number of patients required. There should be sufficient power to detect the targeted treatment effect in the biomarker-positive subgroup, but the biomarker-negative subgroup is likely to be descriptive. The “biomarker strategy” design, whereby patients are randomized to be treated uniformly versus treated differentially according to biomarker status, is statistically inefficient as it includes the biomarker-negative patients who are treated identically in both arms (66). For those reasons, it is not preferred.

More complicated designs allow for integrating treatment and biomarker evaluation (70) in phase III trials. One example allows for testing the treatment effect separately in the marker positive and negative subgroups, as well as in the total patient population. The alpha (defined as the probability of incorrectly rejecting the null hypothesis) is split between the comparison in the marker-positive subgroup and that in the over-all population (for example, 0.015 and 0.010, respectively), and the comparison is conducted first in the marker-positive subgroup. If this test is positive, then the comparison is conducted at total alpha (0.025) in the marker-negative subgroup. On the other hand, if it is negative, then the comparison is conducted in the over-all population at the remainder of the alpha (0.010). This approach allows for testing in both marker subgroups as well as including a back-up comparison in the overall population, in case the biomarker turns out to be nonpredictive. An inappropriate variant of this approach is to conduct the comparison in the total population, rather than in the marker negative subgroup, if the comparison is positive in the marker-positive subgroup. The serious flaw in this is that it can result in generalizing the recommendation for treatment to the marker-negative subgroup, with high probability, if the effect is sufficiently strong in the marker-positive subgroup or if that subgroup is a sufficiently large fraction of the whole, even if there is no treatment benefit in the marker negative subgroup (70).

Biomarkers can also be incorporated in phase II trials (65, 68), to initially evaluate both the efficacy of the treatment and the potentially predictive value of the biomarker, to determine which phase III trial design is most appropriate. Freidlin and colleagues (68) give precise rules for distinguishing among the following:

  1. The treatment is ineffective in the total population and it is ineffective in the biomarker-positive subgroup, in particular, and, therefore, it should not be tested further.

  2. The treatment is promising overall, with no predictive value to the biomarker, and should be tested in a phase III trial with no biomarker incorporation.

  3. The treatment is promising in the biomarker-positive subgroup only and should be tested in a phase III trial so restricted.

  4. The treatment is promising, but the predictive value of the biomarker is unclear, and should be tested in a biomarker stratified phase III trial.

The above designs represent suggestions of different scenarios for biomarker-driven investigations. Given the identified challenges in procuring tissue for biomarker research, it is critical that selected endpoints be part of focused research questions. Biomarker investigations in phase 0 and phase I trials are beyond the scope of this article. While phase 0 trials generally have pharmacodynamic primary endpoints and phase I trials may have expansion cohorts to address pharmacodynamic secondary or exploratory endpoints, neither, in general, address the relation of potential biomarkers to clinical outcomes.

While immunotherapy shows real promise for some patients, responses do not occur in most patients, although those who achieve responses, especially complete or near complete responses, may enjoy long-term durable remissions. It is thus of critical importance to develop biomarkers that predict which patients are most likely to benefit. These efforts are enthusiastically supported by patient communities as long as they are involved in this process to ensure that future guidelines produce useful results for patients (72, 73).

Which biomarkers are patients most interested in?

Overall, predictive biomarkers contain the most promise for patients, whether they predict treatment response, adverse event profiles, or long-term remission. The most important biomarker, however, depends on each patient's situation. For instance, patients who do not have cancer are most interested in prognostic biomarkers of risk and actions they can take to avoid cancer. Patients with early-stage cancers value prevention of recurrence, so predictive markers that help them identify valuable treatments are a priority. For patients with metastatic disease, pharmacodynamic measures of response to treatment, and validated predictive markers of response are most important. Common to all of these scenarios is a desire for tests that provide accurate, reproducible results that help them make treatment decisions. Unfortunately, few immuno-oncology biomarkers are available that meet these needs of patients. Currently available biomarkers that direct optimal selection and sequencing of therapy, such as PD-L1 expression, are imperfect at best given the variability of the assay as well as the inability to account for the complex response relationships of the TME. While an accurate, reliable biomarker is of highest priority in development, the value of blood-based biomarkers that can reliably affect what is happening in the TME, compared with tumor that may be invasive to procure and limited by heterogeneity, is apparent.

When a biomarker recommends treatments of similar efficacy, measures of toxicity are of paramount importance to patients. For this reason, there is a clear need to develop biomarkers that predict toxicity in this setting. Patient-reported outcomes (PRO) and quality-of-life (QOL) studies may yield important insights and are discussed further by Anagnostou and colleagues in this series (74). For example, a recent analysis of patient-reported outcomes from the CheckMate-067 trial showed differences in QOL, global health, and symptom burden between patients with melanoma treated with nivolumab, ipilimumab, or the combination, which allows us to consider whether patients are willing to tolerate significant toxicities if survival is higher (75). In this setting, biomarkers of toxicity are of great value as they could predict the development of side effects for that patient group and can be used to guide treatment choices.

Finally, financial burden is an important consideration and biomarker tests that predict benefit from a specific treatment will help patient avoid the cost of therapy that is not likely to be effective (76). This makes it even more important to integrate biomarkers with patient outcome data to assess true value to patients (77). It is imperative to recall that impressive clinical trial results, if financially out of reach for the vast majority of patients, cannot be considered a true success.

With cancer immunotherapy likely to become a cornerstone of therapy for multiple cancer types, the importance of reliable biomarkers cannot be understated. It is important to remember that the development of immuno-oncology biomarkers is in its infancy, with few adequately validated assays that predict response to therapy. Given the complex and expensive nature of immuno-oncology biomarker discovery, focused research questions involving comprehensive tissue collections with carefully selected endpoints are a priority. In addition, incorporating patient preferences and perspectives is critical in the search for biomarkers of value. The future of immuno-oncology biomarker discovery shows great promise as early studies reveal novel biomarkers that are predictive of response and toxicity and these areas of research are being vigorously pursued.

J.M. Mehnert reports receiving other commercial research support from AstraZeneca, EMD Serono, Immunocore, Incyte, Macrogenics, and Merck and is a consultant/advisory board member for EMD Serono, Genentech, Merck, and Pfizer. A.M. Monjazeb reports receiving other commercial research support from Genentech, Incyte, Merck, and Transgene. No potential conflicts of interest were disclosed by the other authors.

J.M. Mehnert is supported by the National Cancer Institute (UM1CA186716).

1.
Brahmer
JR
,
Tykodi
SS
,
Chow
LQM
,
Hwu
W-J
,
Topalian
SL
,
Hwu
P
, et al
Safety and activity of anti–PD-L1 antibody in patients with advanced cancer
.
N Engl J Med
2012
;
366
:
2455
65
.
2.
Hodi
FS
,
O'Day
SJ
,
McDermott
DF
,
Weber
RW
,
Sosman
JA
,
Haanen
JB
, et al
Improved survival with ipilimumab in patients with metastatic melanoma
.
N Engl J Med
2010
;
363
:
711
23
.
3.
Topalian
SL
,
Hodi
FS
,
Brahmer
JR
,
Gettinger
SN
,
Smith
DC
,
McDermott
DF
, et al
Safety, activity, and immune correlates of anti-PD-1 antibody in cancer
.
N Engl J Med
2012
;
366
:
2443
54
.
4.
McDermott
DF
,
Drake
CG
,
Sznol
M
,
Choueiri
TK
,
Powderly
JD
,
Smith
DC
, et al
Survival, durable response, and long-term safety in patients with previously treated advanced renal cell carcinoma receiving nivolumab
.
J Clin Oncol
2015
;
33
:
2013
20
.
5.
Gettinger
SN
,
Horn
L
,
Gandhi
L
,
Spigel
DR
,
Antonia
SJ
,
Rizvi
NA
, et al
Overall survival and long-term safety of nivolumab (Anti-Programmed Death 1 Antibody, BMS-936558, ONO-4538) in patients with previously treated advanced non-small-cell lung cancer
.
J Clin Oncol
2015
;
33
:
2004
12
.
6.
Rizvi
NA
,
Hellmann
MD
,
Snyder
A
,
Kvistborg
P
,
Makarov
V
,
Havel
JJ
, et al
Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer
.
Science
2015
;
348
:
124
8
.
7.
Ansell
SM
,
Lesokhin
AM
,
Borrello
I
,
Halwani
A
,
Scott
EC
,
Gutierrez
M
, et al
PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma
.
N Engl J Med
2015
;
372
:
311
9
.
8.
Nghiem
PT
,
Bhatia
S
,
Lipson
EJ
,
Kudchadkar
RR
,
Miller
NJ
,
Annamalai
L
, et al
PD-1 blockade with pembrolizumab in advanced merkel-cell carcinoma
.
N Engl J Med
2016
;
374
:
2542
52
.
9.
Chen
R
,
Zinzani
PL
,
Fanale
MA
,
Armand
P
,
Johnson
NA
,
Brice
P
, et al
Phase II study of the efficacy and safety of pembrolizumab for relapsed/refractory classic hodgkin lymphoma
.
J Clin Oncol
2017
;
35
:
2125
32
.
10.
Reck
M
,
Rodríguez-Abreu
D
,
Robinson
AG
,
Hui
R
,
Csőszi
T
,
Fülöp
A
, et al
Pembrolizumab versus chemotherapy for PD-L1–positive non–small-cell lung cancer
.
N Engl J Med
2016
;
375
:
1823
33
.
11.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
JJ
,
Cowey
CL
,
Lao
CD
, et al
Combined nivolumab and ipilimumab or monotherapy in untreated melanoma
.
N Engl J Med
2015
;
373
:
23
34
.
12.
Ott
PA
,
Bang
Y-J
,
Berton-Rigaud
D
,
Elez
E
,
Pishvaian
MJ
,
Rugo
HS
, et al
Safety and antitumor activity of pembrolizumab in advanced programmed death ligand 1-positive endometrial cancer: results from the KEYNOTE-028 study
.
J Clin Oncol
2017 May 10
.
[Epub ahead of print]
.
13.
Hamanishi
J
,
Mandai
M
,
Ikeda
T
,
Minami
M
,
Kawaguchi
A
,
Murayama
T
, et al
Safety and antitumor activity of anti-PD-1 antibody, nivolumab, in patients with platinum-resistant ovarian cancer
.
J Clin Oncol
2015
;
33
:
4015
22
.
14.
Galon
J
,
Pagès
F
,
Marincola
FM
,
Angell
HK
,
Thurin
M
,
Lugli
A
, et al
Cancer classification using the Immunoscore: a worldwide task force
.
J Transl Med
2012
;
10
:
205
.
15.
Bindea
G
,
Mlecnik
B
,
Tosolini
M
,
Kirilovsky
A
,
Waldner
M
,
Obenauf
AC
, et al
Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer
.
Immunity
2013
;
39
:
782
95
.
16.
Spranger
S
,
Spaapen
RM
,
Zha
Y
,
Williams
J
,
Meng
Y
,
Ha
TT
, et al
Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells
.
Sci Transl Med
2013
;
5
:
200ra116
.
17.
Hegde
PS
,
Karanikas
V
,
Evers
S
. 
The Where, the When, and the How of immune monitoring for cancer immunotherapies in the era of checkpoint inhibition
.
Clin Cancer Res
2016
;
22
:
1865
74
.
18.
Ribas
A
,
Robert
C
,
Hodi
FS
,
Wolchok
JD
,
Joshua
AM
,
Hwu
W-J
, et al
Association of response to programmed death receptor 1 (PD-1) blockade with pembrolizumab (MK-3475) with an interferon-inflammatory immune gene signature.
J Clin Oncol
33
:
15s
, 
2015
(
suppl; abstr 3001
).
19.
Hamid
O
,
Schmidt
H
,
Nissan
A
,
Ridolfi
L
,
Aamdal
S
,
Hansson
J
, et al
A prospective phase II trial exploring the association between tumor microenvironment biomarkers and clinical activity of ipilimumab in advanced melanoma
.
J Transl Med
2011
;
9
:
204
.
20.
Tumeh
PC
,
Harview
CL
,
Yearley
JH
,
Shintaku
IP
,
Taylor
EJM
,
Robert
L
, et al
PD-1 blockade induces responses by inhibiting adaptive immune resistance
.
Nature
2014
;
515
:
568
71
.
21.
Salgado
R
,
Denkert
C
,
Demaria
S
,
Sirtaine
N
,
Klauschen
F
,
Pruneri
G
, et al
The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014
.
Ann Oncol
2015
;
26
:
259
71
.
22.
Garon
EB
,
Rizvi
NA
,
Hui
R
,
Leighl
N
,
Balmanoukian
AS
,
Eder
JP
, et al
Pembrolizumab for the treatment of non–small-cell lung cancer
.
N Engl J Med
2015
;
372
:
2018
28
.
23.
Jorgensen
JT
. 
Companion diagnostic assays for PD-1/PD-L1 checkpoint inhibitors in NSCLC
.
Expert Rev Mol Diagn
2016
;
16
:
131
3
.
24.
Lipson
EJ
,
Forde
PM
,
Hammers
H-J
,
Emens
LA
,
Taube
JM
,
Topalian
SL
. 
Antagonists of PD-1 and PD-L1 in cancer treatment
.
Semin Oncol
2015
;
42
:
587
600
.
25.
Herbst
RS
,
Soria
J-C
,
Kowanetz
M
,
Fine
GD
,
Hamid
O
,
Gordon
MS
, et al
Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients
.
Nature
2014
;
515
:
563
7
.
26.
Taube
JM
,
Klein
A
,
Brahmer
JR
,
Xu
H
,
Pan
X
,
Kim
JH
, et al
Association of PD-1, PD-1 Ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy
.
Clin Cancer Res
2014
;
20
:
5064
74
.
27.
Borghaei
H
,
Paz-Ares
L
,
Horn
L
,
Spigel
DR
,
Steins
M
,
Ready
NE
, et al
Nivolumab versus docetaxel in advanced nonsquamous non–small-cell lung cancer
.
N Engl J Med
2015
;
373
:
1627
39
.
28.
Zhang
Y
,
Chen
L
,
EJ
L
,
PM
F
,
HJ
H
,
LA
E
, et al
Classification of advanced human cancers based on tumor immunity in the MicroEnvironment (TIME) for cancer immunotherapy
.
JAMA Oncol
2016
;
2
:
1403
.
29.
Gajewski
TF
,
Schreiber
H
,
Fu
Y-X
. 
Innate and adaptive immune cells in the tumor microenvironment
.
Nat Immunol
2013
;
14
:
1014
22
.
30.
Day
D
,
Monjazeb
AM
,
Sharon
E
,
Ivy
SP
,
Rubin
EH
,
Rosner
GL
, et al
From famine to feast: developing early-phase combination immunotherapy trials wisely
.
Clin Cancer Res
2017
;
23
:
4980
91
.
31.
Sanmamed
MF
,
Chen
L
. 
Inducible expression of B7-H1 (PD-L1) and its selective role in tumor site immune modulation
.
Cancer J
2014
;
20
:
256
61
.
32.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
33.
Mehnert
JM
,
Panda
A
,
Zhong
H
,
Hirshfield
K
,
Damare
S
,
Lane
K
, et al
Immune activation and response to pembrolizumab in POLE-mutant endometrial cancer
.
J Clin Invest
2016
;
126
:
2334
40
.
34.
Johnson
DB
,
Frampton
GM
,
Rioth
MJ
,
Yusko
E
,
Xu
Y
,
Guo
X
, et al
Targeted next generation sequencing identifies markers of response to PD-1 blockade
.
Cancer Immunol Res
2016
;
4
:
959
67
.
35.
Snyder
A
,
Makarov
V
,
Merghoub
T
,
Yuan
J
,
Zaretsky
JM
,
Desrichard
A
, et al
Genetic basis for clinical response to CTLA-4 blockade in melanoma
.
N Engl J Med
2014
;
371
:
2189
99
.
36.
Van Allen
EM
,
Miao
D
,
Schilling
B
,
Shukla
SA
,
Blank
C
,
Zimmer
L
, et al
Genomic correlates of response to CTLA-4 blockade in metastatic melanoma
.
Science
2015
;
350
:
207
11
.
37.
McGranahan
N
,
Furness
AJS
,
Rosenthal
R
,
Ramskov
S
,
Lyngaa
R
,
Saini
SK
, et al
Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade
.
Science
2016
;
351
:
1463
9
.
38.
Bigley
AB
,
Rezvani
K
,
Shah
N
,
Sekine
T
,
Balneger
N
,
Pistillo
M
, et al
Latent cytomegalovirus infection enhances anti-tumour cytotoxicity through accumulation of NKG2C+ NK cells in healthy humans
.
Clin Exp Immunol
2016
;
185
:
239
51
.
39.
Green
MR
,
Rodig
S
,
Juszczynski
P
,
Ouyang
J
,
Sinha
P
,
O'Donnell
E
, et al
Constitutive AP-1 activity and EBV infection induce PD-L1 in Hodgkin lymphomas and posttransplant lymphoproliferative disorders: implications for targeted therapy
.
Clin Cancer Res
2012
;
18
:
1611
8
.
40.
Panda
A
,
Mehnert
JM
,
Hirshfield
KM
,
Riedlinger
G
,
Damare
S
,
Saunders
T
, et al
Immune activation and prolonged benefit to avelumab (anti-PD-L1) therapy in a patient with metastatic EBV+ gastric cancer
.
J Immunother Cancer
2016
;
4
(
Suppl 1
):
P95
.
41.
Roemer
MGM
,
Advani
RH
,
Ligon
AH
,
Natkunam
Y
,
Redd
RA
,
Homer
H
, et al
PD-L1 and PD-L2 genetic alterations define classical hodgkin lymphoma and predict outcome
.
J Clin Oncol
2016
;
34
:
2690
7
.
42.
Oguejiofor
K
,
Galletta-Williams
H
,
Dovedi
SJ
,
Roberts
DL
,
Stern
PL
,
West
CML
. 
Distinct patterns of infiltrating CD8+ T cells in HPV+ and CD68 macrophages in HPV-oropharyngeal squamous cell carcinomas are associated with better clinical outcome but PD-L1 expression is not prognostic
.
Oncotarget
2017
;
8
:
14416
27
.
43.
Pitt
JM
,
Vétizou
M
,
Waldschmitt
N
,
Kroemer
G
,
Chamaillard
M
,
Boneca
IG
, et al
Fine-tuning cancer immunotherapy: optimizing the gut microbiome
.
Cancer Res
2016
;
76
:
4602
7
.
44.
Dubin
K
,
Callahan
MK
,
Ren
B
,
Khanin
R
,
Viale
A
,
Ling
L
, et al
Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis
.
Nat Commun
2016
;
7
:
10391
.
45.
Sivan
A
,
Corrales
L
,
Hubert
N
,
Williams
JB
,
Aquino-Michaels
K
,
Earley
ZM
, et al
Commensal Bifidobacterium promotes antitumor immunity and facilitates anti–PD-L1 efficacy
.
Science
2015
;
350
:
1084
9
.
46.
Wargo
JA
,
Gopalakrishnan
V
,
Spencer
C
,
Karpinets
T
,
Reuben
A
,
Andrews
MC
, et al
Association of the diversity and composition of the gut microbiome with responses and survival (PFS) in metastatic melanoma (MM) patients (pts) on anti-PD-1 therapy
.
J Clin Oncol
2017
;
35
:
3008
.
47.
Ferris
RL
,
Blumenschein
G
 Jr
,
Harrington
K
,
Fayette
J
,
Guigay
J
,
Colevas
AD
. 
Evaluation of oral microbiome profiling as a response biomarker in squamous cell carcinoma of the head and neck: analyses from CheckMate 141 [abstract]
.
In
:
Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1–5
;
Washington, DC. Philadelphia (PA)
:
AACR
; 
2017
.
Abstract nr CT022
.
48.
Gopalakrishnan
V
,
Spencer
C
,
Reuben
A
,
Karpinets
T
,
Hutchinson
D
,
Hoffman
K
. 
Association of diversity and composition of the gut microbiome with differential responses to PD-1 based therapy in patients with metastatic melanoma
.
J Clin Oncol
2017
.
49.
Twyman-Saint Victor
C
,
Rech
AJ
,
Maity
A
,
Rengan
R
,
Pauken
KE
,
Stelekati
E
, et al
Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer
.
Nature
2015
;
520
:
373
7
.
50.
Nyberg
K
. 
Leveraging immune biomarkers in the blood to guide immunotherapy—current application and future prospects ASCO-SITC clinical immuno-oncology symposium
. 
2017
.
51.
Schuurhuis
DH
,
Verdijk
P
,
Schreibelt
G
,
Aarntzen
EH
,
Scharenborg
N
,
de Boer
A
, et al
In situ expression of tumor antigens by messenger RNA-electroporated dendritic cells in lymph nodes of melanoma patients
.
Cancer Res
2009
;
69
:
2927
34
.
52.
Tavare
R
,
Escuin-Ordinas
H
,
Mok
S
,
McCracken
MN
,
Zettlitz
KA
,
Salazar
FB
, et al
An effective immuno-PET imaging method to monitor CD8-dependent responses to immunotherapy
.
Cancer Res
2016
;
76
:
73
82
.
53.
McShane
LM
,
Cavenagh
MM
,
Lively
TG
,
Eberhard
DA
,
Bigbee
WL
,
Williams
PM
, et al
Criteria for the use of omics-based predictors in clinical trials
.
Nature
2013
;
502
:
317
20
.
54.
Haanen
J
. 
Controversy of the day - biomarkers in immunotherapy: where are we now?
European Society for Medical Oncology Daily Reporter
. 
2016
.
55.
Marrero
A
,
Lawrence
S
,
Wilsker
D
,
Voth
AR
,
Kinders
RJ
. 
Translating pharmacodynamic biomarkers from bench to bedside: analytical validation and fit-for-purpose studies to qualify multiplex immunofluorescent assays for use on clinical core biopsy specimens
.
Semin Oncol
2016
;
43
:
453
63
.
56.
Baik
CS
,
Rubin
EH
,
Forde
PM
,
Mehnert
JM
,
Collyar
D
,
Butler
MO
, et al
Immuno-oncology clinical trial design: limitations, challenges, and opportunities
.
Clin Cancer Res
2017
;
23
:
4992
5002
.
57.
International Melanoma Tissue Bank Consortium (IMTBC)
.
Available from
: https://www.aimatmelanoma.org/melanoma-research/international- melanoma-tissue-bank/.
58.
Zaretsky
JM
,
Garcia-Diaz
A
,
Shin
DS
,
Escuin-Ordinas
H
,
Hugo
W
,
Hu-Lieskovan
S
, et al
Mutations associated with acquired resistance to PD-1 blockade in melanoma
.
N Engl J Med
2016
;
375
:
819
29
.
59.
Parchment
RE
,
Doroshow
JH
. 
Theory and practice of clinical pharmacodynamics in oncology drug development
.
Semin Oncol
2016
;
43
:
427
35
.
60.
Kohrt
HE
,
Tumeh
PC
,
Benson
D
,
Bhardwaj
N
,
Brody
J
,
Formenti
S
, et al
Immunodynamics: a cancer immunotherapy trials network review of immune monitoring in immuno-oncology clinical trials
.
J Immunother Cancer
2016
;
4
:
15
.
61.
Gibney
GT
,
Weiner
LM
,
Atkins
MB
. 
Predictive biomarkers for checkpoint inhibitor-based immunotherapy
.
Lancet Oncol
2016
;
17
:
e542
51
.
62.
Prost
S
,
Kishen
REB
,
Kluth
DC
,
Bellamy
COC
,
Pinaud
F
,
Michalet
X
, et al
Working with commercially available quantum dots for immunofluorescence on tissue sections
.
PLoS One
2016
;
11
:
e0163856
.
63.
Singer
DS
,
Jacks
T
,
Jaffee
E
. 
A U.S. “Cancer Moonshot” to accelerate cancer research
.
Science
2016
;
353
.
64.
Pepke
S
,
Ver Steeg
G
. 
Comprehensive discovery of subsample gene expression components by information explanation: therapeutic implications in cancer
.
BMC Med Genomics
2017
;
10
:
12
.
65.
McShane
LM
,
Hunsberger
S
,
Adjei
AA
. 
Effective incorporation of biomarkers into phase II trials
.
Clin Cancer Res
2009
;
15
:
1898
905
.
66.
Freidlin
B
,
McShane
LM
,
Korn
EL
. 
Randomized clinical trials with biomarkers: design issues
.
J Natl Cancer Inst
2010
;
102
:
152
60
.
67.
Freidlin
B
,
Korn
EL
. 
Biomarker-adaptive clinical trial designs
.
Pharmacogenomics
2010
;
11
:
1679
82
.
68.
Freidlin
B
,
McShane
LM
,
Polley
M-YC
,
Korn
EL
. 
Randomized phase II trial designs with biomarkers
.
J Clin Oncol
2012
;
30
:
3304
9
.
69.
Polley
M-YC
,
Freidlin
B
,
Korn
EL
,
Conley
BA
,
Abrams
JS
,
McShane
LM
. 
Statistical and practical considerations for clinical evaluation of predictive biomarkers
.
J Natl Cancer Inst
2013
;
105
:
1677
83
.
70.
Freidlin
B
,
Sun
Z
,
Gray
R
,
Korn
EL
. 
Phase III clinical trials that integrate treatment and biomarker evaluation
.
J Clin Oncol
2013
;
31
:
3158
61
.
71.
Freidlin
B
,
Korn
EL
. 
Biomarker enrichment strategies: matching trial design to biomarker credentials
.
Nat Rev Clin Oncol
2013
;
11
:
81
90
.
72.
U.S. Food and Drug Administration
. 
A blueprint proposal for companion diagnostic comparability [Internet]
; 
2015
.
73.
Brennan
Z
. 
Multiple endpoints in clinical trials: FDA issues draft guidance [Internet]
; 
2017
.
74.
Anagnostou
V
,
Yarchoan
M
,
Hansen
AR
,
Wang
H
,
Verde
F
,
Sharon
E
, et al
Immuno-oncology trial endpoints: capturing clinically meaningful activity
.
Clin Cancer Res
2017
;
23
:
4959
69
.
75.
Schadendorf
D
,
Long
G
,
Larkin
J
. 
Patient reported outcomes (PROs) from a phase 3 study of nivolumab (NIVO) alone or combined with ipilimumab (IPI) versus IPI in patients with advanced melanoma. CheckMate 067
.
Present Soc Melanoma Res
2015
Int Congr [Internet]
.
76.
National Academies of Sciences
.
Biomarker tests for molecularly targeted therapies [Internet]
.
Graig
LA
,
Phillips
JK
,
Moses
HL
,
editors
.
Washington, D.C.
:
National Academies Press
; 
2016
.
77.
Blank
CU
,
Haanen
JB
,
Ribas
A
,
Schumacher
TN
. 
The “cancer immunogram.”
Science
2016
;
352
.