Download as pdf or txt
Download as pdf or txt
You are on page 1of 21

REVIEW

The Next Decade of Immune


Checkpoint Therapy
Padmanee Sharma1,2,3, Bilal A. Siddiqui1, Swetha Anandhan1, Shalini S. Yadav3, Sumit K. Subudhi1,
Jianjun Gao1, Sangeeta Goswami1,2, and James P. Allison2,3

ABSTRACT Immune checkpoint therapy (ICT) can provide durable clinical responses and
improve overall survival. However, only subsets of patients with specific tumor
types respond to ICT. Thus, significant challenges remain, including understanding pathways of resist-
ance, optimizing patient selection, improving management of immune-related adverse events, and
identifying rational therapeutic combinations. These challenges will need a focused approach encom-
passing both clinical and basic research, with the integration of reverse translational studies. This
integrated approach will lead to identification of potential targets for subsequent clinical trials, which
will guide decisions as we develop novel combination strategies to maximize efficacy and minimize
toxicities for patients.

Significance: ICTs induce durable antitumor responses for subsets of patients with cancer. Recent
evidence suggests that rational combinatorial strategies can improve response by overcoming primary
and adaptive resistance mechanisms, although these may carry an increased risk of immune-mediated
toxicities. This review surveys the current understanding of mechanisms of response and resistance to
ICTs and active areas of investigation, and proposes a path forward to improving efficacy and minimiz-
ing toxicities through better patient selection and rational combinations.

INTRODUCTION Despite the unprecedented durable clinical responses


observed in subsets of patients, most patients do not respond,
Metastatic cancers remain incurable in most patients as
and some patients develop resistance to therapy after initial
conventional therapies that target tumor cells usually can-
response. Furthermore, ICTs can result in life-threatening
not provide a durable response. The discovery of immune
toxicities, known as immune-related adverse events (irAE).
checkpoints that regulate immune responses transformed
Therefore, research studies are ongoing to understand path-
cancer care, offering long-term clinical responses and the
ways of resistance to ICT and mechanisms of irAEs. In this
possibility of cure in many more patients with metastatic
review, we outline the fundamentals of ICT and the current
cancer (1). In 2011, the FDA approved the first immune
state of ICT in the clinic. We propose that development of
checkpoint targeting agent, ipilimumab—a mAb targeting
effective ICT combinations will require a reverse translational
CTLA4—which opened the field of immune checkpoint ther-
approach, in which hypotheses are generated from in-depth
apy (ICT). Subsequently, mAbs blocking other checkpoints
immune monitoring studies of patients’ samples and then
such as PD-1 or PD-L1 have received FDA approvals to treat
tested in appropriate preclinical models, thereby yielding
a number of tumor types alone and in combination with
important insights to guide rational therapeutic strategies
other agents.
in subsequent clinical trials. The last ten years provided an
understanding of how T cells can be targeted with anti-
CTLA4 and anti–PD-1/PD-L1 to provide clinical benefit.
1
Department of Genitourinary Medical Oncology, The University of Texas Bolstered by the latest technologies, in the next decade we will
MD Anderson Cancer Center, Houston, Texas. 2Department of Immunology, better understand how to combine ICT with agents targeting
The University of Texas MD Anderson Cancer Center, Houston, Texas. 3The
Immunotherapy Platform, The University of Texas MD Anderson Cancer,
multiple other cell subsets and pathways in the innate and
Center, Houston, Texas. adaptive immune system. The future of ICT offers ongoing
Corresponding Author: Padmanee Sharma, Genitourinary Medical Oncol- promise to improve outcomes in patients with cancer.
ogy and Immunology, The University of Texas MD Anderson Cancer Center,
1515 Holcombe Boulevard, Houston, TX 77030. Phone: 713-552-3980;
E-mail: padsharma@mdanderson.org BACKGROUND
Cancer Discov 2021;11:838–57 T cells are the soldiers of the immune response. Activation
doi: 10.1158/2159-8290.CD-20-1680 of T cells requires a coordinated and stepwise process involv-
©2021 American Association for Cancer Research. ing two primary signals (Fig. 1; ref. 2). T cells express the T-cell

838 | CANCER DISCOVERY 10TH ANNIVERSARY ISSUE April 2021 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
The Next Decade of Immune Checkpoint Therapy REVIEW

A T-cell activation B T-cell inhibition

PD-L1/L2
PD-1

MHCII-Ag TCR MHCII-Ag TCR


CTLA4
CD28
CD28

B7 B7

Cytokine release

C T-cell reactivation using immune


checkpoint antibodies

PD-L1/L2
PD-1

Other coinhibitory
molecules
MHCII-Ag TCR
CD28
CTLA4

B7

Cytokine release

Figure 1.  Regulation of T-cell activation. A, T-cell activation requires both signal 1, TCR engagement with the MHC–peptide antigen complex (MHC-
Ag) on an APC or a target cell, and signal 2, interaction of the costimulatory receptor CD28 on the T cell with costimulatory B7 molecules (CD80/CD86).
B, In response to T-cell activation, the immune checkpoints CTLA4 and PD-1 are upregulated on the T cell and bind to B7 and PD-L1/L2, respectively, to
inhibit T-cell activation. C, Immune checkpoint antibodies targeting CTLA4 or PD-1/PD-L1 block these inhibitory interactions, reactivating T cells.

receptor (TCR), which signals via the CD3 complex upon the T-bet for Th1 cells, BCL6 for Tfh, RORγ for Th17 cells, and
interaction of the TCR with MHC plus its cognate peptide anti- FOXP3 for Tregs. Although CD4 T cells are known as helper
gen on antigen-presenting cells (APC). T cells also constitutively cells, accumulating evidence suggests a cytotoxic function
express a costimulatory receptor, CD28, which binds to the demonstrated by certain CD4 T cells (4). CD8 T cells are typi-
B7 family of costimulatory molecules that are predominantly cally regarded as a uniform population with cytotoxic func-
expressed on professional APCs. To activate or turn T cells “on,” tion; however, recent studies have highlighted the diversity of
both the TCR signal via CD3 (signal 1) and the CD28 costimu- the CD8 T-cell population (5–9).
latory signal via B7 interactions (signal 2) are required (Fig. 1A). In general, Th1 CD4 cells and CD8 T cells are the effector
Interaction of APCs with T cells that bear the specific TCR for a cells responsible for driving antitumor immune responses.
given antigen leads to activation of T cells and T cell–mediated However, these immune responses are controlled by multiple
responses. T cells then differentiate and clonally expand, with mechanisms. Upon T-cell activation via the TCR and CD28,
subsequent formation of memory T cells (3). T cells proliferate and produce cytokines, but also upregulate
CD4 and CD8 T cells constitute the majority of T cells in inhibitory molecules that attenuate T-cell activation (Fig. 1B).
the immune system. CD4 T cells differentiate into a variety Therefore, T cells have limited time to function before they are
of subtypes: Th1, Th2, Th17, follicular Th (Tfh), T regulatory reined in to prevent damage to normal cells. Multiple inhibi-
(Treg), and Th9 cells. These subsets have distinct transcrip- tory mechanisms exist, both cell-intrinsic and cell-extrinsic,
tion factors that regulate their function and cytokine profiles, to control T-cell responses. The signals that control antitu-
which affect immune responses that range from effector cell mor immunity and immune suppression are comprised of a
responses for tumor rejection to regulatory cell responses to tightly regulated process that forms the yin and yang of the
suppress effector cells. These transcription factors include immune response.

April 2021 CANCER DISCOVERY 10TH ANNIVERSARY ISSUE | 839

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
REVIEW Sharma et al.

CTLA4-Mediated Negative Costimulation tumor activity of CTLA4 blockade demonstrated the contri-
CTLA4 is a canonical inhibitory molecule that is expressed bution of both the effector T-cell and Treg compartments in
immediately following engagement of TCR, with peak preclinical models (22, 23). Of note, although CTLA4 block-
expression around 48 to 72 hours following T-cell activa- ade was shown to cause selective depletion of intratumoral
tion. It bears structural homology to CD28 and binds to Tregs through antibody-dependent cell-mediated cytotoxic-
B7-1 (CD80) and B7-2 (CD86) molecules on APCs with ity (ADCC) in murine models and ex vivo experiments on
higher affinity than CD28, leading to competitive inhibition human tumor samples (24–26), subsequent studies compar-
of costimulatory CD28 signaling, thereby dampening T-cell ing pretreatment and posttreatment tumor samples from
signaling (10, 11). In early studies, an anti-CTLA4 antibody patients who received anti-CTLA4 therapy, either as treme-
prevented CTLA4 from engaging B7, leading to prolonged limumab (IgG2 antibody) or as ipilimumab (IgG1 anti-
T-cell responses and eradication of cancer cells (1). Simi- body), did not observe a decrease in intratumoral Tregs after
larly, the in vivo administration of anti-CTLA4 antibodies therapy (27). It is possible that the expression of specific Fc
enhanced the antitumor effect of tumor-infiltrating T cells, receptors on APCs or other cell types may be limited in some
leading to regression of established tumors and long-lived human tumors, thereby affecting Treg depletion by anti-
immunity in tumor models (1). CTLA4 therapy in patients with cancer. In future studies,
Antibody blockade of CTLA4 was proposed as a way to it may be important to ascertain the expression of specific
enhance T-cell responses in the setting of cancer. By removing Fc receptors within the tumor microenvironment (TME) to
the brakes from T cells, it was hypothesized that T cells would determine whether ICT antibodies will mediate ADCC to
have a longer time to function, which would enable tumor affect Treg frequency. In general, anti-CTLA4 expands CD4
rejection. Anti-CTLA4 opened an entire new field termed effector T cells (Teff) with subsequent increase in the Teff/
“ICT.” The success of anti-CTLA4 led to the identification of Treg ratio in responding tumors (22).
other pathways that regulate T-cell responses, including both Anti–PD-1/PD-L1 antibodies predominantly affect exhausted
costimulatory and coinhibitory pathways (Fig. 1C; refs. 1, 12). CD8 T cells. Anti–PD-1/PD-L1 also does not appear to expand
clonal diversity nor promote T-cell trafficking into tumors. Pre-
PD-1–Mediated Negative Costimulation clinical studies have shown that concurrent targeting of CTLA4
and PD-1/PD-L1 can improve therapeutic efficacy when com-
The primary biological function of PD-1 is to maintain
pared with each alone (28–30). mAbs targeting CTLA4, PD-1,
peripheral tolerance. PD-1 engages with its ligands PD-L1 and
and PD-L1 have formed the foundation of effective ICT in
PD-L2, widely expressed on nonlymphoid tissues, primarily
patients and are now in widespread clinical use across a number
dampening T-cell activation in the periphery. PD-1 interacts
of tumor types (31–34).
with an SHP2 tyrosine phosphatase domain, leading to dephos-
phorylation of signaling molecules downstream of the TCR
and inhibiting TCR-mediated activation of IL2 production and CURRENT ICTs IN THE CLINIC
T-cell proliferation (13). Therefore, whereas CTLA4 inhibits sig-
There are now more than 50 FDA approvals for ICTs in
nal 2 (CD28-B7), PD-1/PD-L1 inhibits signal 1 (TCR).
human cancers, which are briefly discussed in this section
PD-L1 expression has been described on a variety of tumor
and summarized in Table 1. The clinically approved ICTs
types, and is directly correlated with poor prognosis in several
have been comprehensively reviewed in other sources (35, 36).
cancers (14, 15). Preclinical models demonstrated that PD-1
blockade results in tumor rejection through reinvigoration Anti-CTLA4 Monotherapy
of exhausted CD8 T cells that express PD-1, LAG3, and TIM3
Ipilimumab (anti-CTLA4) was the first ICT to gain FDA
and expansion of CD8 T cells, a hallmark of response to anti–
approval in 2011 for the treatment of metastatic melanoma,
PD-1 therapy (16).
on the basis of phase III trials demonstrating improvements
in overall survival compared with vaccine or chemotherapy
Distinct Mechanisms of Action of CTLA4 and PD-1 (37, 38). Initial studies were challenging because of the para-
Although both CTLA4 and PD-1 are inhibitory molecules doxical phenomenon of pseudoprogression, in which patients
that attenuate T-cell activation, they have different mecha- initially experienced tumor growth before subsequent regres-
nisms of action, and inhibition of CTLA4 and PD-1 leads to sion. The benefit of ICT would not have been adequately
distinct immune responses. Anti-CTLA4 primarily functions captured by traditional metrics of tumor measurement
in T-cell priming and expands clonal diversity. Anti-CTLA4 (39–41). Survival therefore proved to be the most impor-
predominantly affects CD4 T cells, with multiple studies tant indicator of ICT benefit. Indeed, approximately 20% of
identifying increased frequency of a Th1 subset of CD4 T patients who received ipilimumab achieved long-term sur-
cells that express inducible costimulator (ICOS) as a result of vival greater than three years, with survival of 10 years noted
anti-CTLA4 therapy (17–19). Increased frequency of ICOS+ for some patients (38, 42). Although ipilimumab remains
CD4 T cells in peripheral blood acts as a pharmacodynamic the only anti-CTLA4 therapy to have approval in many coun-
biomarker of anti-CTLA4 therapy (20), whereas increased tries, including FDA approval, other mAbs are being stud-
frequency of ICOS+ CD4 T cells in tumor tissues, possibly in ied, including tremelimumab and quavonlimab (43). New
the context of tertiary lymphoid structures, tends to correlate forms of anti-CTLA4 therapies are also under development,
with improved clinical outcomes with ICT (21). Anti-CTLA4 including glycoengineered antibodies that seek to enhance
can also promote T-cell trafficking into immunologically ADCC and prodrug forms with reduced systemic activity to
“cold” tumors. Investigation into the mechanisms of anti­ improve safety (44).

840 | CANCER DISCOVERY 10TH ANNIVERSARY ISSUE April 2021 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
The Next Decade of Immune Checkpoint Therapy REVIEW

Table 1. FDA-approved ICTs

Year of
Agent/target Indication approval
1. Ipilimumab/anti-CTLA4  1. Unresectable or metastatic melanoma 2011
 2. High-risk stage III melanoma after complete resection (adjuvant) 2015
 3. Pediatric patients 12 years and older with unresectable or metastatic melanoma 2017
2. Nivolumab/anti–PD-1  1. Unresectable or metastatic melanoma 2014
 2. Metastatic squamous NSCLC with progression on or after platinum-based chemotherapy 2015
 3. Metastatic nonsquamous NSCLC with progression on or after platinum-based 2015
chemotherapy
 4. Patients with metastatic RCC who have received prior therapy 2015
 5. cHL that has relapsed or progressed after autologous hematopoietic stem cell transplan- 2016
tation and posttransplantation brentuximab vedotin or after ≥3 lines of therapy
 6. Recurrent or metastatic HNSCC with disease progression on or after platinum-based 2016
therapy
 7. Locally advanced or metastatic UC with disease progression during or following platinum- 2017
based chemotherapy
 8. Adult and pediatric patients with MSI-H or dMMR metastatic colorectal cancer that has 2017
progressed following chemotherapy
 9. HCC previously treated with sorafenib 2017
10. Melanoma with lymph node involvement (adjuvant) or metastatic disease following 2017
complete resection
11. Unresectable advanced, recurrent, or metastatic esophageal squamous cell carcinoma 2020
after prior fluoropyrimidine- and platinum-based chemotherapy
12. Advanced RCC, first line in combination with cabozantinib 2021
3. Pembrolizumab/anti–PD-1  1. Advanced or unresectable melanoma that no longer responds to other drugs 2014
 2. Metastatic NSCLC with PD-L1–positive tumors and disease progression after other 2015
treatments
 3. First-line treatment of unresectable or metastatic melanoma 2015
 4. Recurrent or metastatic HNSCC with disease progression on or after platinum-based 2016
chemotherapy
 5. First-line treatment of metastatic NSCLC, with high PD-L1 expression and no EGFR or 2016
ALK genomic tumor alterations
 6. Adult and pediatric refractory cHL or disease relapse after three or more prior lines of 2017
therapy (extended in 2020 to adult patients with relapsed or refractory cHL and pediatric
patients with refractory cHL or cHL relapsed after two or more lines of therapy)
 7. First-line treatment of metastatic nonsquamous NSCLC, irrespective of PD-L1 expres- 2017
sion (in combination with carboplatin/pemetrexed)
 8. Locally advanced or metastatic UC with disease progression on or after platinum-based 2017
chemotherapy or within 12 months of neoadjuvant or adjuvant platinum chemotherapy
or patients who are not eligible for cisplatin-containing chemotherapy and whose tumors
express PD-L1 (CPS ≥10), or in patients who are not eligible for any platinum-containing
chemotherapy regardless of PD-L1 status
 9. Adult and pediatric unresectable or metastatic solid tumors that are MSI-H or dMMR 2017
10. Recurrent locally advanced or metastatic gastric or gastroesophageal junction adeno- 2017
carcinoma, with PD-L1–positive tumors and disease progression on or after two or more
prior lines of therapy
11. Recurrent or metastatic cervical cancer, with tumor PD-L1 CPS ≥ 1 and disease progres- 2018
sion on or after prior chemotherapy
12. Primary mediastinal B-cell lymphoma, refractory disease, or relapse after two or more 2018
lines of therapy
13. Metastatic, nonsquamous NSCLC (in combination with pemetrexed and cisplatin/ 2018
carboplatin)
14. Metastatic, squamous NSCLC (in combination with carboplatin and paclitaxel or 2018
nab-paclitaxel)

(continued)

April 2021 CANCER DISCOVERY 10TH ANNIVERSARY ISSUE | 841

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
REVIEW Sharma et al.

Table 1. FDA-approved ICTs (Continued)

Year of
Agent/target Indication approval
15. HCC, following treatment with sorafenib 2018
16. Merkel cell carcinoma, recurrent locally advanced or metastatic 2018
17. Melanoma with lymph node involvement following complete resection 2019
18. Locally advanced or metastatic NSCLC with PD-L1 TPS ≥ 1% 2019
19. Advanced RCC, first line (in combination with axitinib) 2019
20. HNSCC, first-line treatment for metastatic or unresectable recurrent disease with tumor 2019
PD-L1 CPS ≥ 1
21. HNSCC, first-line treatment for metastatic or unresectable recurrent disease (in combi- 2019
nation with platinum and fluorouracil)
22. SCLC, metastatic, with disease progression on or after platinum-based chemotherapy 2019
and at least 1 other prior line of therapy
23. Esophageal cancer, recurrent locally advanced or metastatic squamous cell, with tumor 2019
PD-L1 CPS ≥ 10 and disease progression after one or more prior lines of systemic therapy
24. Endometrial carcinoma, advanced, non–MSI-H/dMMR with disease progression after 2019
prior systemic therapy and not candidate for curative surgery or radiation (in combination
with lenvatinib)
25. UC, high-risk, BCG-unresponsive, non–muscle invasive with carcinoma in situ with or 2020
without papillary tumors, ineligible for or declined cystectomy
26. Recurrent or metastatic cutaneous squamous cell carcinoma not curable by surgery or 2020
radiation
27. Unresectable or metastatic TMB-high (≥10 mut/Mb) solid tumors that have progressed 2020
on prior treatment with no satisfactory alternative treatment options.
28. Unresectable or metastatic MSI-H or dMMR colorectal cancer, first line 2020
29. Locally recurrent unresectable or metastatic triple-negative breast cancer with tumor 2020
PD-L1 CPS ≥ 10 (in combination with chemotherapy)
4. Ipilimumab + nivolumab/  1. BRAFV600 wild-type unresectable or metastatic melanoma 2015
anti-CTLA4 + anti–PD-1  2. BRAFV600 wild-type and BRAFV600 mutation–positive unresectable or metastatic 2016
melanoma
 3. Poor/intermediate risk previously untreated advanced RCC 2018
 4. Previously treated MSI-H/dMMR colorectal cancer 2018
 5. HCC following treatment with sorafenib 2020
 6. Metastatic NSCLC with PD-L1 ≥1% or in combination with two cycles of platinum-doublet 2020
chemotherapy regardless of PD-L1 status
 7. Unresectable malignant pleural mesothelioma 2020
5. Durvalumab/anti–PD-L1  1. Unresectable stage III NSCLC with nonprogressive disease following concurrent 2018
platinum-based chemotherapy and radiotherapy
 2. SCLC, extensive stage, first line (in combination with etoposide and carboplatin/cisplatin) 2020
6. Atezolizumab/anti–PD-L1  1. Locally advanced or metastatic UC, with disease progression during or following 2016
platinum-based chemotherapy, either before or after surgery
 2. Metastatic NSCLC with disease progression during or following platinum-based chemo- 2016
therapy, and progression on an FDA-approved targeted therapy if the tumor has EGFR or
ALK gene abnormalities
 3. Locally advanced or metastatic UC not eligible for cisplatin chemotherapy whose tumors 2017
express PD-L1
 4. First-line, metastatic, nonsquamous SCLC without EGFR or ALK genomic alterations 2018
(in combination with bevacizumab, paclitaxel, and carboplatin)
 5. SCLC, extensive stage, first line (in combination with etoposide and carboplatin) 2019
 6. Unresectable locally advanced or metastatic triple-negative breast cancer with PD-L1 2019
≥1% (in combination with nab-paclitaxel)
 7. Metastatic NSCLC without EGFR or ALK genomic alterations (in combination with 2019
carboplatin/nab-paclitaxel)

842 | CANCER DISCOVERY 10TH ANNIVERSARY ISSUE April 2021 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
The Next Decade of Immune Checkpoint Therapy REVIEW

Table 1. FDA-approved ICTs (Continued)

Year of
Agent/target Indication approval
 8. Metastatic NSCLC, first line, PD-L1 high (tumor cells ≥50% or immune cells ≥10%) 2020
without EGFR or ALK genomic alterations
 9. Metastatic or unresectable HCC, first line (in combination with bevacizumab) 2020
10. BRAFV600 mutation–positive unresectable or metastatic melanoma in combination with 2020
cobimetinib and vemurafenib
7. Avelumab/anti–PD-L1  1. Adult and pediatric patients with metastatic Merkel cell carcinoma, including those who 2017
have not received prior chemotherapy
 2. Locally advanced or metastatic UC with disease progression during or following platinum- 2017
based chemotherapy
 3. Advanced RCC, first line (in combination with axitinib) 2019
 4. Maintenance treatment for locally advanced for metastatic UC that has not progressed 2020
with first-line platinum-containing chemotherapy
8. Cemiplimab/anti–PD-1  1. Metastatic or locally advanced cutaneous squamous cell carcinoma 2018

Abbreviations: BCG, Bacillus Calmette–Guérin; cHL, classic Hodgkin lymphoma; CPS, combined positive score; dMMR, mismatch-repair deficiency;
HCC, hepatocellular carcinoma; HNSCC, head and neck squamous cell carcinoma; MSI-H, microsatellite instability-high; NSCLC, non–small cell lung
cancer; RCC, renal cell carcinoma; SCLC, small cell lung cancer; TPS, tumor proportion score; TMB, tumor mutational burden; UC, urothelial carcinoma.
Updated as of February 1, 2021.

Anti–PD-1/PD-L1 Monotherapy either monotherapy, in different cancer types including mel-


In 2014, the first PD-1 inhibitors were approved for the anoma (54, 55), hepatocellular carcinoma (56), renal cell
treatment of unresectable or metastatic melanoma (45, 46). carcinoma (RCC; refs. 57, 58), NSCLC (59), malignant pleural
The PD-1/PD-L1 inhibitors are now approved across mul- mesothelioma (60), and colorectal cancer (MSI-H/dMMR;
tiple tumor types, including skin, genitourinary, lung, head ref. 61). Long-term follow-up has demonstrated durable sur-
and neck, breast, lymphoma, gynecologic, and gastrointes- vival benefit for a subset of patients with melanoma and RCC,
tinal cancers. These approvals are summarized in Table 1. and combination therapy with ICT is under active investiga-
The PD-1 mAbs include nivolumab (fully human IgG4), tion for a number of tumor types (55, 58).
pembrolizumab (humanized IgG4), and cemiplimab (fully
human IgG4). The PD-L1 mAbs include durvalumab (fully Combination of ICT with Other Therapeutic Agents
human IgG1κ), atezolizumab (humanized IgG1), and ave- The quest to improve response rates has also prompted
lumab (fully human IgG1). Anti–PD-1 therapies have also combinations with other established cancer drugs, for exam-
received approval for treatment of patients with cancer who ple, chemotherapy, which is hypothesized to kill immuno-
are diagnosed with tumors that have microsatellite instability– suppressive cells in the TME while increasing exposure to
high (MSI-H)/mismatch-repair deficiency (dMMR; refs. 47, tumor antigens via the induction of tumor cell death, thereby
48) or high tumor mutational burden (TMB-high) regardless enhancing recognition and elimination of tumor cells by the
of tumor type (49). In addition, anti–PD-1/PD-L1 therapies host immune system (62). Toward this end, combinations of
were also approved by the FDA as maintenance therapy, which ICTs with concurrent and/or sequential chemotherapy are
is administered after initial treatment of locally advanced or under extensive investigation, with phase III clinical data dem-
metastatic disease. Clinical benefit with maintenance ICT onstrating overall survival and/or progression-free survival
in specific settings was demonstrated in patients with non– benefit in NSCLC (63–66), small cell lung cancer (51, 52), tri-
small cell lung cancer (NSCLC; ref. 50), extensive-stage small ple-negative breast cancer (67), bladder cancer (53), and head
cell lung cancer (51, 52), and bladder cancer (53), leading to and neck squamous cell carcinoma (68). However, the impact
FDA approvals in these tumor types. of different chemotherapy agents on the immune response
within the TME has not been fully elucidated. Similarly,
Combination of Anti-CTLA4 and Anti–PD-1/PD-L1 improved clinical responses have been demonstrated with
Despite the promising responses with ICT monotherapies, the combination of ICT and targeted therapy. Combinations
these responses are observed in only 20% to 30% of treated of VEGF-targeted agents with anti–PD-1 have demonstrated
patients. Therefore, combination therapy was pursued in clinical benefits in endometrial carcinoma (pembrolizumab
the clinic based on preclinical data demonstrating that anti- plus lenvatinib; refs. 69, 70), RCC (pembrolizumab plus axi-
CTLA4 and anti–PD-1/PD-L1 function through distinct tinib; nivolumab plus cabozantinib; avelumab plus axitinib),
pathways, and combined inhibition enhanced antitumor and hepatocellular carcinoma (atezolizumab plus bevaci-
responses in murine models (28). zumab; refs. 71–75). Recently, the addition of atezolizumab
Multiple clinical trials with combined blockade of CTLA4 to targeted therapy was also shown to improve progression-
and PD-1/PD-L1 demonstrated higher response rates than free survival in BRAF-mutated melanoma (76).

April 2021 CANCER DISCOVERY 10TH ANNIVERSARY ISSUE | 843

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
REVIEW Sharma et al.

Although these combination therapies offer the possi- IMPROVING CLINICAL OUTCOMES WITH
bility of improving cancer-specific outcomes, they remain RATIONAL ICT COMBINATIONS
limited to specific tumor types and disease settings. For
the field of cancer immunotherapy to move forward suc- To obtain a greater understanding of mechanisms of
cessfully, with improved clinical benefit for many more response and resistance to ICT, we propose that reverse trans-
patients, we will require rational combination therapies, lational studies on individual agents be undertaken, whereby
which should be based on a reasonable understanding of each agent can be studied in small cohorts of patients, with
the mechanism of action of each agent and its impact on longitudinal samples collected and analyzed thoroughly to
immune responses. generate hypotheses regarding immunologic mechanisms
that can be tested in appropriate preclinical models, and with
Immune-Related Adverse Events translation of the preclinical data to new clinical trials. These
As combination strategies are pursued, there is an increased data sets may provide relevant information that can help with
risk of development of irAEs. Mitigation strategies, therefore, the design of rational clinical trials as well as avoid the pit-
will require a deeper mechanistic understanding of the toxici- falls of failed clinical trials. We have adopted this integrated
ties. These irAEs are distinct from the predictable side effects strategy to understand the pathways regulating immune
associated with conventional cancer therapies. The irAEs responses and to guide rational combinatorial strategies, as
can affect any organ, most commonly skin, gastrointestinal outlined below.
tract, and endocrine system (including the thyroid, adrenal,
and pituitary glands). The severity of irAEs can range from Addressing Primary and Adaptive
mild to even fatal. Clinical guidelines for the management Resistance Mechanisms
of irAEs have been reviewed elsewhere (77). Although guide- Prostate cancer is considered a “cold” tumor due to the fact
lines recommend high-dose corticosteroids for severe irAEs, that it has few mutations, as compared with “hot” tumors
often with prolonged tapers, this approach can be associated such as melanoma and NSCLC. As a result of a low number
with serious iatrogenic side effects, highlighting the need for of mutations, and therefore few antigens for T-cell recogni-
mechanistically informed, steroid-sparing immunosuppres- tion, as noted by scarce tumor-infiltrating T cells, prostate
sive approaches. Early recognition and treatment of irAEs is cancer has been predominantly resistant to treatment with
essential to prevent progression to severe events, and major ICT (100–102). The mechanisms by which prostate tumors
efforts are under way to develop biomarkers for earlier pre- exclude T cells or fail to elicit robust infiltration by T cells
dictions of irAEs, including blood-based biomarkers such as represent primary resistance mechanisms to ICT. Without
CD8 T-cell clonal expansion, serum autoantibodies (reflective tumor-infiltrating T cells, the blockade of a T cell–inhibitory
of B-cell response), and technology-based approaches with pathway such as PD-1/PD-L1 is unlikely to provide clinical
patient symptom reporting (78–81). benefit. For example, a recent phase III clinical trial of the
Published data indicate that nonfatal irAEs may be asso- androgen receptor antagonist enzalutamide plus anti–PD-L1
ciated with improved outcomes in patients with cancer failed to meet its primary endpoint of improved overall
(82–96). Therefore, the development of strategies that can survival (103). Previous studies on human prostate cancers
enable patients to continue on ICT, including the use of demonstrated few tumor-infiltrating T cells, with minimal
selective immunosuppressive agents, will be important as the to no expression of PD-1 or PD-L1, and lack of significant
field of ICT moves forward. For example, the recent develop- clinical benefit with anti–PD-1 monotherapy in patients with
ment of a murine model of myocarditis, one of the most metastatic prostate cancer (104, 105). Therefore, the phase III
feared complications of ICT, provided a rationale for the use combination study would have benefited from a smaller clini-
of CTLA4-Ig (abatacept), which blocks T-cell costimulation, cal trial to truly determine whether enzalutamide was capa-
although an open question remains as to whether this will ble of increasing tumor-infiltrating T cells with subsequent
interfere with antitumor immunity (97, 98). Similarly, in increase in PD-1 and PD-L1 expression within the TME (106).
a study of patients with melanoma who developed colitis Similarly, a phase III combination study of anti-CTLA4 plus
after ICT, single-cell RNA sequencing (scRNA-seq) and TCR radiotherapy in patients with metastatic prostate cancer also
sequencing identified tissue-resident colitis-associated CD8 failed to meet its primary endpoint (100, 107). The combina-
T cells, consistent with the clinical observation that colitis tion was conducted without understanding the immunologic
symptoms tend to occur early after initiation of ICT, and impact of anti-CTLA4 or radiotherapy on the prostate TME,
also provided support for the use of the α4β7 antibody ved- including sites of metastatic disease, which are predominantly
olizumab to decrease inflammatory immune responses in bone metastases in patients with metastatic prostate cancer.
ICT-associated colitis (99). To address the immunologic impact of anti-CTLA4 on the
Overall, as multiple mechanisms related to irAEs become prostate TME, a small clinical trial evaluated pretreatment
clear, including both T cell– and B cell–mediated pathways, a and posttreatment tumor samples, which demonstrated that
major question will be to address whether the mechanisms of anti-CTLA4 was capable of increasing tumor-infiltrating T
irAEs can successfully be decoupled from those of antitumor cells (105). However, anti-CTLA4 also led to an increase in
efficacy. Future ICT clinical trials may include components compensatory inhibitory mechanisms, also known as adaptive
of selective, prophylactic immunosuppression for high-risk, resistance mechanisms, including increased frequency of cells
biomarker-selected patients to prevent severe irAEs, or pos- expressing PD-1/PD-L1 and VISTA within the TME (105).
sibly include administration of targeted immunosuppression VISTA is an inhibitory checkpoint that is highly expressed on
to enable continuation of ICT in patients. myeloid cells [including macrophages, dendritic cells (DC),

844 | CANCER DISCOVERY 10TH ANNIVERSARY ISSUE April 2021 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
The Next Decade of Immune Checkpoint Therapy REVIEW

monocytes, neutrophils], and moderately expressed on T cells knockout mice showed improved survival with ICT compared
(108). This yin and yang of the immune response, where ther- with control mice, demonstrating a direct role for CD73
apy may drive antitumor immune responses but also increase (112). These series of studies helped in the identification of
immunosuppressive mechanisms, is an important concept to CD73 as a specific immunotherapeutic target to improve
take into consideration as new immunotherapeutic strategies antitumor immune responses to ICT in GBM, generating a
are developed. combination strategy for a future clinical trial in GBM.
The data from this small clinical trial generated hypotheses Other myeloid-specific targets, including chemokine recep-
regarding combination therapies with anti-CTLA4 plus anti– tors (CXCR4, CCR2, and CSF1R), intracellular mediators
PD-1 and/or anti-VISTA. These hypotheses were tested in (IDO1 and PI3Kϒ), and proangiogenic agents (VEGF receptors,
preclinical studies, and the data led to a new study with anti- semaphorin 4D and angiopoietin-2), are also being explored
CTLA4 plus anti–PD-1 (109). The new combination clinical in an attempt to limit the function of immunosuppressive
trial elicited antitumor responses in some patients, but these (M2) macrophages while promoting the function of M1 mac-
responses were predominantly in patients with soft-tissue rophages, which facilitate antitumor immune responses (113).
metastases as opposed to bone metastases (109). Immune The differentiation of macrophages into M1 or M2 subsets
monitoring data of posttreatment tumor samples collected is a complex biological process that is being investigated.
from patients indicated that soft-tissue metastases were infil- Many different signals, including cytokine and epigenetic
trated by Th1 cells, whereas bone metastases were infiltrated signals, appear to play a role in driving macrophage differ-
by Th17 cells (110). The increased Th1 responses provided an entiation. Interestingly, the process of phagocytosis, which
explanation for why soft-tissue metastases responded better is required for clearing tumor cells, also seems to play a
than bone metastases to ICT. role in driving M2 differentiation in an attempt to suppress
But why did ICT drive Th1 responses in soft-tissue metas- immune responses and avoid further cell death (114), indicat-
tases and Th17 responses in bone metastases? A preclinical ing the complexity of how immune responses are regulated.
model was designed to answer this question. Data from these Detailed understanding of the signals and biological pro-
experiments demonstrated high levels of IL6 and TGFβ in cesses involved in the differentiation of macrophages will be
bone metastases as opposed to soft-tissue metastases, which needed to effectively target these cells.
are important cytokines for skewing T-cell response toward
Th17 instead of Th1 (110). These preclinical data are now Epigenetics and Immune Response
being used to develop a new combination therapy clinical As outlined previously, the functional and phenotypic
trial with ICT plus an agent to target TGFβ. states of immune subpopulations play an important role
On the basis of these data, the concept of organ-specific in determining response to ICT. Cellular plasticity is often
immune microenvironments will need to be addressed as the driven by epigenetic mechanisms. Although the role of the
field of ICT moves forward in the next decade. Treatment of epigenetic machinery in regulating cancer cell plasticity has
patients with metastatic disease will need to take into consid- been characterized (115), we lack mechanistic insight into
eration whether certain organs, such as bone and liver, have the epigenetic regulation of immune subsets, especially in the
distinct immune microenvironments that will require differ- context of ICT.
ent immunotherapeutic strategies (111). It was demonstrated that ipilimumab induces the expres-
sion of EZH2, a key epigenetic enzyme that plays an impor-
Immunosuppressive Myeloid Cells tant role in cellular plasticity (115–118). CD28 signaling leads
The immune microenvironment is unique to each tumor to increased EZH2 expression on T cells (119). Anti-CTLA4
type and plays a critical role in response and resistance to increases CD28 signaling, thereby leading to increased EZH2
ICT. Therefore, it is important to understand the composi- expression on T cells. On the basis of these observations, a
tion and phenotypic states of intratumoral immune cells series of preclinical studies were designed that identified a
of different tumor types to develop a tumor-specific ICT potential role for EZH2 in driving adaptive resistance to anti-
strategy. Analysis of the immune microenvironment of five CTLA4 therapy. The combination of EZH2 inhibition with
different tumor types, including those that respond relatively anti-CTLA4 therapy led to improved survival in the preclinical
well to ICT and those that do not, revealed that ICT-sensitive studies (116). These studies formed the basis for a clinical trial
and ICT-resistant tumor types have distinct immune micro- of the rational combination of an EZH1/2 inhibitor (DS3201)
environments. Interaction of tumor cells with the immune with ipilimumab in patients with genitourinary malignancies
microenvironment guides the constitution and phenotypes with primary resistance to ICT (NCT04388852). This repre-
of immune subpopulations. sents the tip of the iceberg in terms of epigenetic regulation of
Immunologically cold tumors, such as prostate cancer and immune cells, providing the foundation for the combination
glioblastoma multiforme (GBM), have few tumor-infiltrating of epigenetic modulators with ICT (120).
T cells but higher frequency of immune-suppressive myeloid It is clear that we currently have the tools at our disposal to
cells. Comparison of tumor-infiltrating lymphocytes (TIL) interrogate human immune responses in great depth and we
from GBM and multiple other tumor types led to the iden- will need to take advantage of these tools, and combine the
tification of a GBM-specific CD73hi myeloid cell population, human data with appropriate preclinical models, to identify
which persists even after treatment with anti–PD-1 (112). rational targets for combination strategies that will improve
CD73 is an extracellular ectonucleotidase that plays a role in clinical outcomes for patients. Patient selection will also play
the adenosine immunosuppressive pathway, inhibiting T-cell an important part in developing effective treatments, and
activation and proliferation. Preclinical studies with CD73 we will need to consider multiple factors, including both

April 2021 CANCER DISCOVERY 10TH ANNIVERSARY ISSUE | 845

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
REVIEW Sharma et al.

rs Im
ke m
ar un
m • TMB
• T cells
e-
io • Neoantigens


Teff/Treg ratio
IFNγ signature

sp
b
• PD-L1 expression • B-cell signature
c

ec
• DNA damage pathways CD103+ DCs
si

ifi
• MSI status
rin

• TLS

cb
• WNT/β-catenin • CXCL13
int

• IFN signaling mutations • PD-L1 expression

iom
(JAK )
or-

• M1 and M2 cells (CD73+, VISTA+)


• ARID1A mutation
Tum

arke
• PTEN loss
PD-L1
Neoantigens
Monocytes
Tumor cells

rs
Stromal cells Macrophages
TMB Cytokines
TLS
T cells
Driver mutations Biomarkers DCs
affecting
response and
resistance
to ICT

C
om
bi
• TMB and PD-L1
s e
na
to r
• TMB and IFNγ signature
p on
es
• ARID1A and CXCL13
ial
bio m i ct r
arkers to pred

Figure 2.  Biomarkers of response and resistance to ICT. Tumor cell–specific and immune cell–specific biomarkers associated with response and
resistance to ICT, and combinatorial biomarkers that may predict response to ICT. DC, dendritic cell; MSI, microsatellite instability; Teff, effector T cell;
TLS, tertiary lymphoid structure; TMB, tumor mutational burden; Treg, regulatory T cell.

tumor-related factors and immune-related factors, to identify understanding of determinants of antitumor immunity has
appropriate populations. improved, we appreciate that single biomarkers in isolation
are insufficient. Combinatorial biomarker strategies that cap-
ture attributes of the host and tumor–immune ecosystem will
PREDICTIVE BIOMARKERS FOR OPTIMAL
be essential (Fig. 2; refs. 121–124).
PATIENT SELECTION
One of the major challenges in the field of ICT is the lack of TMB and Neoantigen Load as
a robust predictive biomarker for optimal patient selection. Predictive Biomarkers
Traditionally, biomarkers have focused primarily on tumor- High TMB is associated with response to ICT in numerous
intrinsic factors or single immune-specific markers. As our cancer types, most notably melanoma and NSCLC (49). High

846 | CANCER DISCOVERY 10TH ANNIVERSARY ISSUE April 2021 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
The Next Decade of Immune Checkpoint Therapy REVIEW

TMB may contribute to the formation of neoantigens that PD-L1 Expression as a Predictive Biomarker
are recognized by the immune system (125, 126). On a tissue- IHC evaluation of PD-L1 expression on tumor and/or
agnostic basis, anti–PD-1 (pembrolizumab) received FDA immune cells is frequently performed to select patients for
approval for the treatment of advanced pediatric and adult treatment with anti–PD-1/PD-L1 antibodies. A correla-
solid tumors that have high TMB (≥10 mutations/megabase). tion between PD-L1 expression on tumor cells and clinical
This is a promising step on which to build, and future stud- response is seen in certain circumstances (e.g., in NSCLC);
ies on TMB as a predictive biomarker will address threshold however, in other cases, substantial responses can be observed
validation as well as provide a deeper understanding of the in some patients with PD-L1–negative tumors, which high-
immunogenicity of different mutational patterns (127). lights the difficulty of using PD-L1 as a single biomarker
Although high TMB led to FDA approval, it should be (34, 143, 144). PD-L1 expression is dynamic due to multiple
noted that certain tumor types with moderate TMB carry rel- factors, including T-cell responses and IFNγ signaling. There-
atively high response rates to ICT, such as clear cell RCC (128, fore, variations in the timing of biopsy of the tumor as well
129). In addition, low TMB does not preclude generation of as the site of biopsy may influence whether PD-L1 expres-
effective antigen responses, nor does high TMB guarantee sion is detected or not. Although a range of assays have been
response to ICT (130–132). For example, it was shown that approved, there remains significant variation in antibodies,
anti-CTLA4 therapy can induce effective neoantigen-specific tissue handling techniques, and thresholds, which limits
T-cell responses in patients with prostate cancer, even in the the uniform benefit of PD-L1 expression as an isolated bio-
background of low TMB (131). Therefore, one approach to marker, and efforts are under way to integrate PD-L1 expres-
enhance antigen-specific T-cell responses may be the combi- sion with other biomarkers (described below; ref. 127).
nation of ICT with personalized neoantigen vaccines, which
have been shown to modulate T cells in the TME and enhance
responses to ICT, including in both immunologically “cold” TILs and Exclusion of TILs as Biomarkers of
tumors such as glioblastoma and “hot” tumors such as mela- Response and Resistance
noma (133–135). As vaccine therapies evolve to encompass TILs have a long history in cancer immunotherapy, with
appropriate formulations, adjuvants, and delivery methods demonstration in the 1980s that adoptive transfer of TILs
that can elicit effective antitumor immune responses, it may derived from patients’ melanoma tumors can lead to cancer
be possible to take advantage of tumor mutations to generate regression (145). It was later shown that TILs, including CD8
neoantigen vaccines in combination with ICT for the devel- T cells, correlated with clinical outcomes in multiple tumor
opment of successful therapeutic strategies. types, including ovarian cancer and bladder cancer (146,
147). Subsequent research on the correlates of response of
DNA Damage Repair Pathways: MSI-H/dMMR and anti-CTLA4 therapy demonstrated that the extent of ICT-
Homologous Repair Defects induced tumor necrosis was directly related to the ratio of
There are fundamental links between DNA damage and intratumoral CD8 T cells to FOXP3+ Tregs (148). TILs are
immunogenicity. Tumors with DNA damage repair (DDR) therefore under active investigation as a biomarker to guide
defects, exemplified by MSI-H/dMMR status, were predicted selection of patients for treatment with ICT, although signifi-
to have high rates of neoantigen formation due to their cant variability exists in definitions and thresholds (149, 150).
high mutational load (approximately 10-fold compared with In a prospective study, the AMADEUS trial (NCT03651271,
that of MMR-proficient tumors) and therefore higher rates ongoing) was designed to evaluate the benefit of classifying
of response to ICT (136). An initial study in patients with tumors into immunologically “hot” and “cold” tumors based
colorectal cancer demonstrated significantly higher rates of on CD8+ T-cell density (≥15% and <15%, respectively), with
response to pembrolizumab in dMMR patients compared anti–PD-1 (nivolumab) monotherapy administered as treat-
with MMR-proficient patients (47), a finding that was later ment for “hot” tumors, whereas anti-CTLA4 (ipilimumab)
extended to multiple tumor types (48). These data led to plus anti–PD-1 (nivolumab) combination is administered
the tumor-agnostic approval of pembrolizumab for MSI-H/ as treatment for “cold” tumors in an attempt to take advan-
dMMR solid tumors (48). tage of the ability of anti-CTLA4 to drive T cells into “cold”
Additionally, homologous repair defects, such as patho- tumors. In addition, the Immunoscore is an approach that
genic mutations in BRCA1/2, can lead to enhanced immune aims to standardize and quantify CD3 and CD8 T cells infil-
response by multiple pathways including the release of cyto- trating and surrounding a tumor (149, 151). However, due
solic DNA, promotion of type I IFN signaling, and upreg- to variability in immune cell infiltration in different tumor
ulation of PD-L1 (137–139). In a study of patients with types, its applicability across cancers is challenging. Multiple
urothelial carcinoma, higher response rates to anti–PD-1/ studies indicate that the location of T cells in the tumor
PD-L1 antibodies were observed in patients with known (whether at the invasive margins or in tumor parenchyma)
or likely deleterious DDR alterations (140). Emerging evi- and the phenotype of the T cells (whether cells are memory-
dence indicates that specific DDR mutations have differential like, exhausted, and/or activated) will need to be taken into
effects on the tumor–immune microenvironment, and there- consideration (152–154).
fore mutations will need to be selected carefully as candidate In the same vein, T-cell exclusion is a mechanism of resist-
predictive biomarkers (141). In all, an integrated approach ance to ICT, exemplified by “cold” tumors such as prostate
incorporating events in specific driver genes and mutational cancer and pancreatic cancer, which have few T cells in the
signatures will be critical to develop additional genetic bio- TME. Certain oncogenic pathways may enable tumors to
markers of response (142). exploit this mechanism of immune evasion. For example,

April 2021 CANCER DISCOVERY 10TH ANNIVERSARY ISSUE | 847

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
REVIEW Sharma et al.

tumor cell–intrinsic WNT/β-catenin activation has been be essential to understand which cells are interacting within
shown to lead to exclusion of T cells within the TME (123, the TLS, and how they differ from the typical structures
155, 156). In addition, PTEN loss is also associated with within lymph nodes. Finally, because TLS appear to be asso-
increased signaling through the PI3K–AKT pathway and ciated with responses to ICT across multiple tumor types, it
correlated with exclusion of CD8 T cells and poor clinical will be important to define the specific mechanisms driving
responses to ICT (124). Thus, patient selection for treatment TLS formation to determine whether these mechanisms can
and strategies to overcome T-cell exclusion in the TME will be exploited to induce the formation of TLS in otherwise
need to be considered in future ICT trials. nonresponding tumors.

IFNg Gene Signature as a Predictive Biomarker Combinatorial Biomarkers


IFNγ is a key cytokine produced by activated T cells, as Although many of the biomarkers described above, includ-
well as natural killer (NK) and NK T cells, and is critical ing tumor-intrinsic and host-specific variables, correlate with
for effective antitumor immunity. Preclinical work using response to ICTs, many of the studies have been retrospective
IFNγ-blocking antibody and murine models lacking essential and need further validation through prospective clinical tri-
IFNγ signaling molecules demonstrated a key role for IFNγ als. Furthermore, there is a critical need to bridge preclini-
in immune surveillance and tumor rejection (157, 158). In cal mechanistic studies with clinical investigations and vice
addition, knockdown of IFNγ-receptor 1 led to impaired versa to develop reproducible predictive biomarkers. There
antitumor responses with anti-CTLA4 in a murine model is growing recognition that single biomarker strategies do
of melanoma (159). Consistent with the preclinical data, not incorporate the dynamic and complex interaction of
patients with melanoma harboring genomic defects in IFNγ the tumor and host immune system and that combinatorial
pathway genes did not respond to anti-CTLA4 (160). Also, biomarkers will be essential to optimize patient selection.
patients with melanoma tumors harboring loss-of-function With inherent inter- and intratumor heterogeneity, a complex
mutations in JAK1 and JAK2, downstream IFNγ signaling interplay of diverse cell types, and the continuously evolving
molecules, were resistant to anti–PD-1 therapy, highlighting tumor immune microenvironment, it is clear that the utility
the utility of IFNγ as a potential biomarker. of isolated biomarkers is limited. A combination approach
IFNγ induces a host of target genes that have been used integrating multiple data sets across different platforms from
to develop gene-expression signatures, which correlate with diverse studies will be essential for developing more sensitive
responses to ICT. The gene-expression signatures can be ret- and robust biomarkers of response.
rospectively labeled as “high” or “low” based on median cutoff Recent studies demonstrated that the combination of
values. Multiple studies have reported that tumors identified existing biomarkers has better predictive capacity than single
as having “high” expression of IFNγ-related gene signatures biomarkers. For example, TMB plus a T cell–inflamed gene-
have better clinical responses to ICT (131, 161, 162). However, expression profile or TMB plus PD-L1 expression showed an
the gene panels used to design the IFN-gene signatures were improved prediction performance compared with single bio-
not consistent across these studies. In addition, consistent with markers (130). A recent study of more than 1,000 ICT-treated
the yin and yang concept of immune responses, IFNγ drives patients across seven tumor types showed that a multivari-
expression of proteins such as PD-L1 and IDO1, which may act able predictor of response outperformed TMB alone (169).
to suppress immune responses in the TME. Additional studies Such combinatorial biomarkers, tested in prospective clinical
are ongoing to develop reproducible assays for detection of trials, will be key for optimal patient selection.
IFNγ-related genes and signatures that may be used as a pro- It was also recently demonstrated that two novel biomark-
spective biomarker for selection of patients for ICT. ers correlate with response to anti–PD-1/PD-L1 therapies.
A retrospective analysis identified mutations in ARID1A in
Tertiary Lymphoid Structures as a tumor tissues and expression of the chemokine CXCL13 as
Predictive Biomarker predictive biomarkers for ICT (170). Preclinical studies dem-
Tertiary lymphoid structures (TLS) have recently emerged onstrated association of ARID1A and CXCL13 with antitumor
as biomarkers of response to ICT across several tumor types, immunity in mice (170). Importantly, the combination of
including lung cancer, melanoma, RCC, sarcoma, and urothe- ARID1A mutation plus CXCL13 showed improved predictive
lial cancer (163–167). TLS or TLS-like structures comprised capacity compared with either of the single biomarkers alone
of T cells, B cells, DCs, and other APCs may play a significant (170). This retrospective study highlighted the importance of
role in antitumor T-cell responses, likely due to further prim- integrating tumor mutational status with the immune micro-
ing and activation of T cells within the TME, which may environment to predict responses to ICT, which led to a pro-
represent another important event in the cancer immunity spective clinical trial to test ARID1A mutation plus CXCL13
cycle. In contrast to other biomarkers of response which give as a combinatorial biomarker strategy for patient selection.
a snapshot of a single time point and single cell type, TLS are
dynamic structures comprised of multiple cell types. Multiple
chemokines and cytokines are involved in attracting these cell NEXT STEPS FOR ICT
types and forming the TLS, including CXCL13 (B cells) and The field of ICT has expanded across many different tumor
IL2 (T cells; ref. 168). types, with improved benefit for patients with cancer. In addi-
TLS represent sites of interactions between multi- tion, as the field continues to highlight other pathways that
ple immune cell types and capture an ongoing antitumor can be targeted, there are now a vast number of combinatorial
response (168). Future studies using newer technologies will approaches than can be tested in patients. Therefore, it will

848 | CANCER DISCOVERY 10TH ANNIVERSARY ISSUE April 2021 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
The Next Decade of Immune Checkpoint Therapy REVIEW

Further understanding of mechanisms of


primary and adaptive resistance to ICT

Development of robust predictive biomarkers for


optimal patient selection

Integration of newer technologies to obtain deeper


biological insights The
next
decade
of ICT
Mechanistic and clinical understanding of irAEs

Incorporation of reverse translational strategies to


develop rational combinations

Figure 3.  Future directions in ICT. Figure highlighting the focus areas in the coming decade in the field of ICT that will play a critical role in improving
patient outcome.

be important to efficiently design future studies with atten- clinical trials. The first neoadjuvant ICT trial was conducted
tion to the mechanisms of action of each agent as well as an in 2006 and provided safety data indicating that ICT can
understanding of resistance mechanisms that evolve over be given prior to surgery (17). The study was conducted in
time to control immune responses. The field of ICT is still patients with localized bladder cancer who received anti-
growing, but as we gain knowledge in multiple areas, as out- CTLA4 before proceeding to cystectomy (17). This study
lined below, the field will mature to provide clinical benefit was also the first ICT trial in patients with bladder can-
for even more patients with cancer (Fig. 3). cer, which provided critical data demonstrating antitumor
responses in patients with bladder cancer, thereby leading
Integration of ICT with Other Therapies to large clinical trials with ICT in patients with metastatic
Over the past decade, ICT has become established as the bladder cancer, with subsequent FDA approval. In addi-
fourth pillar of cancer therapy, along with chemotherapy/ tion, the immune monitoring studies performed on the
targeted therapy, radiotherapy, and surgery. We have seen trial highlighted the reverse translational approach and
some success in combining ICT with chemotherapy and identified ICOS+ CD4 T cells as a novel subset of T cells
targeted therapy. The next decade will provide scientific associated with anti-CTLA4 therapy (17, 18, 171, 172). The
insights into integrating ICT with these pillars of cancer care trial also led to the first combination ICT neoadjuvant trial
to benefit more patients. Exciting preclinical data on radio- with anti-CTLA4 plus anti–PD-L1 in patients with bladder
therapy plus ICT should provide insight into translating it to cancer, which demonstrated clinical responses and correla-
clinical practice. Although surgical options in the metastatic tion of TLS with responses (21). Clinical trials with ICT in
setting are extremely limited, we are exploring combinations the neoadjuvant setting have now demonstrated clinical
of ICT with surgery in the metastatic setting. These studies benefit in multiple tumor types, including melanoma (173,
will broaden the field of ICT across all aspects of oncology 174), Merkel cell carcinoma (175), NSCLC (176), MMR-
as a unifying foundation for rational combination strate- deficient colorectal cancer (177), urothelial carcinoma (21,
gies with chemotherapy, targeted therapy, radiotherapy, and 178), and breast cancer (179–181). Additional studies are
surgery. In addition, a number of other potential candi- being planned, especially as investigators consider whether
dates such as other immune checkpoints and costimulatory to implement survival or other clinical endpoints, includ-
receptors, epigenetic modulators, and metabolic targets hold ing pathologic complete responses, which will enable faster
promise for improving the durable clinical responses seen readout and review for possible approval (182–188).
with ICT. Administration of ICT in the neoadjuvant setting enables
T and B cells to have access to the antigens that are present
Moving from Metastatic to Earlier Disease Stages on tumor tissues, which is in contrast to adjuvant studies
Treatment of patients with earlier stages of disease, with where ICT is given after tumor tissues have been removed,
therapy given in either the neoadjuvant or adjuvant set- thereby limiting the amount of tumor antigens that T and
ting, has the potential to prevent disease relapse and lead B cells encounter. Nonetheless, adjuvant ICT has been suc-
to improved survival. Neoadjuvant ICT offers the possibil- cessful in high-risk melanoma, which led to FDA approval
ity of surgical downstaging and provides ample tissue for (189–193). A number of additional studies are currently
examination of an intact TME, potentially yielding insight ongoing and will provide additional insight into the use of
into posttreatment changes that can inform subsequent ICT in the adjuvant setting.

April 2021 CANCER DISCOVERY 10TH ANNIVERSARY ISSUE | 849

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
REVIEW Sharma et al.

Targeting Innate Immune Responses evaluated (207). Both OX40 and 4-1BB/CD137 agonists have
Although much attention has traditionally focused on been tested in clinical trials. Preclinical work with OX40
adaptive immunity, there is growing interest in the role of and 4-1BB showed efficacy; however, a phase Ib study of an
innate immune responses to boost overall antitumor immu- OX40 agonist combined with atezolizumab demonstrated
nity. The innate immune system is composed of NK cells, safety but limited efficacy in patients (208–210). In addition,
dendritic cells, macrophages, monocytes, neutrophils, eosin- phase I studies with CD137-directed mAb urelumab caused
ophils, basophils, and mast cells. Therapeutic approaches significant elevation of liver enzymes (211), and a second
include stimulating innate responses with microbial peptides anti-CD137 antibody, utomilumab, was well tolerated but
or oncolytic viruses, which have previously been shown to had limited efficacy (212). These studies highlight significant
enhance local and systemic antitumor immunity in combina- knowledge gaps regarding the use of agonist antibodies in
tion with ICT and may be able to improve payload to enhance terms of understanding expression of the target proteins,
agonist signals (194–196). The presence of tumor-infiltrating sequencing of the agonist antibody with ICT and choice of
DCs expressing the transcription factor BATF as well as the ICT agent (anti-CTLA4 vs. anti–PD-1/PD-L1). Currently, a
CD103 marker has been shown to be essential for presenta- clinical trial with anti-CTLA4 plus an agonist anti-ICOS
tion of neoantigens and activation of cytotoxic T cells in the antibody is ongoing (NCT03989362) based on preclinical
TME (197, 198). Therefore, approaches that enhance and/or data demonstrating enhanced antitumor immune responses
activate these DCs may improve local antitumor responses. in murine models of melanoma and prostate cancer (172). In
Systemic approaches to stimulating innate immunity, such addition, the ICOS agonist GSK3359609 is also under study
as via agonism of stimulator of interferon response cGAMP in a phase III clinical trial (NCT04128696; ref. 213).
interactor 1 (STING) or Toll-like receptors (TLR), have been The development of immune agonists faces a number
challenging (199–201). Therefore, intratumoral delivery rather of challenges, including target affinity, epitope selection,
than systemic delivery of agents to target innate immune cells receptor occupancy, Fc gamma receptor interactions, anti-
may provide an avenue to ensure presentation of tumor- body isotype, and appropriate in vitro assays to clearly distin-
specific antigens, enable effective T-cell responses, and also guish between an antagonist antibody with activity against
reduce systemic toxicities. Intratumoral agents under inves- a costimulatory molecule expressed on immunosuppressive
tigation in clinical trials include mRNAs encoding CD70, cells versus an agonist antibody with activity against the same
CD40 ligand, and TLR4 (202) and oncolytic viruses such as costimulatory molecule expressed on effector cells (214). As a
talimogene laherparepvec, or T-VEC (203, 204), which is FDA- result, the development of an agonist antibody to target T-cell
approved for melanoma and is being explored in a number responses may require additional experimental steps and
of clinical trials. However, successful DC activation in a sup- careful immune monitoring studies of patients to help design
pressive TME continues to remain a challenge and needs to appropriate combination trials with current ICT agents.
be further investigated. As the field builds on current data with agonist antibodies,
novel approaches are being developed to deliver agonist sig-
Targeting Microbiome to Drive Innate and nals in the TME. For example, bispecific antibodies are being
Adaptive Immune Responses explored, including a bispecific antibody to target CD28 plus
a tumor-associated antigen (NCT03972657; ref. 215). This
Furthermore, as our understanding of host effects on the approach of targeting the costimulatory pathway within the
immune system has grown, it is clear that the gut micro- TME, as opposed to systemic targeting, may provide a safer
biome exerts influence over the effectiveness of ICTs (127). way to provide agonist signals to T cells. As these processes
Responses to ICTs have been correlated with commensal gut are optimized, costimulatory receptor targeting is becoming
flora, and there appear to be substantial differences in the a feasible strategy to enhance the efficacy of ICT.
diversity and composition in patients who respond to ICT
versus nonresponders (205, 206). Challenges in investigating Incorporation of Newer Technologies
the gut microbiome include differences in sampling, analysis In the future, spatiotranscriptomic tools such as Digital
platforms, and population heterogeneity, though these can Spatial Profiling and CO-Detection by indEXing (CODEX)
be addressed with the development of standard sequencing may allow us to decipher the cell types and their inter-
and analytic tools. Manipulation of the gut microbiome, actions within the tumor. In addition, the incorporation
such as through fecal microbiota transplantation, antibiotic of single-cell high-throughput technologies into clinical
therapy, or diet, may serve as an adjunct strategy to enhance and preclinical studies will enable better characterization
responses to ICT. These strategies are still in early stages of cell subsets regulating antitumor responses. Assessing
of being explored but will provide important data that will single-cell transcriptomics, proteomics, and chromatin state
affect treatment with ICT. through scRNA-seq, Cellular Indexing of Transcriptomes and
Epitopes by Sequencing (CITE-seq), and single-cell Assay for
Driving Agonist Signals for Combination Transposase-Accessible Chromatin sequencing (scATAC-seq)
Therapy with ICT will enable identification of cellular identity at a single-cell
On the basis of our understanding of T-cell activation, level. Furthermore, integration of various platforms assisted
there is growing interest in driving agonist signals via costim- by novel bioinformatic tools will allow us to understand the
ulatory receptors. However, clinical experience with a CD28 regulatory pathways governing response and resistance to
superagonist that resulted in severe toxicities has led the field ICT. In addition, integration of longitudinal blood-based
to proceed with caution as additional agonist antibodies are assays such as circulating-tumor DNA (ctDNA; ref. 216),

850 | CANCER DISCOVERY 10TH ANNIVERSARY ISSUE April 2021 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
The Next Decade of Immune Checkpoint Therapy REVIEW

which are easier to implement and less invasive as opposed Oncology, Bristol-Meyers Squibb, Amgen, Inc., and Exelixis, Inc.
to longitudinal tumor biopsies, is under investigation as a outside the submitted work. J.P. Allison reports personal fees from
method to monitor responses to ICT. Jounce Therapeutics, Codiak Biosciences, Achelois, Lava Therapeu-
Additionally, the development of better bioinformatics tics, Lytix, Earli, Phenomics, Dragonfly, Hummingbird, ImaginAb,
Forty-Seven, and Polaris outside the submitted work. No disclosures
algorithms that help with neoantigen prediction will enable
were reported by the other authors.
identification of more relevant therapeutic targets. Progress
in the prediction of shared MHC class I antigens in clinical Received November 23, 2020; revised February 1, 2021; accepted
samples has been exciting. A recent study developed a pipe- February 5, 2021; published first April 2, 2021.
line to determine tumor epitope immunogenicity that might
enable prediction of effective antitumor responses with much
higher accuracy than conventional methods (217). However, References
although developing prediction algorithms for class I anti-
1. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor
gens is necessary, it has recently been shown that MHC class II immunity by CTLA-4 blockade. Science 1996;271:1734–6.
neoantigens are also involved in effective tumor responses 2. Sharma P, Allison JP. The future of immune checkpoint therapy.
and will need to be considered (218). Science 2015;348:56–61.
The successful implementation of these technologies will 3. Smith-Garvin JE, Koretzky GA, Jordan MS. T cell activation. Annu
require close collaboration between computational biologists Rev Immunol 2009;27:591–619.
4. Śledzińska A, Vila de Mucha M, Bergerhoff K, Hotblack A, Demane
and immunologists to avoid simplification of the complex
DF, Ghorani E, et al. Regulatory T cells restrain interleukin-2- and
biological and functional processes of immune responses, Blimp-1-dependent acquisition of cytotoxic function by CD4(+) T
thereby maintaining fidelity to the dynamic nature of cell cells. Immunity 2020;52:151–66.
subsets within the immune system. For example, PD-1 5. Khan O, Giles JR, McDonald S, Manne S, Ngiow SF, Patel KP, et al.
expression on a T cell may represent an “activated” T cell TOX transcriptionally and epigenetically programs CD8(+) T cell
or an “exhausted” T cell. The simple identification of PD-1 exhaustion. Nature 2019;571:211–8.
through a bioinformatics approach may not provide suf- 6. Scott AC, Dündar F, Zumbo P, Chandran SS, Klebanoff CA, Shakiba
M, et al. TOX is a critical regulator of tumour-specific T cell differ-
ficient understanding regarding the complexity of the T-cell entiation. Nature 2019;571:270–4.
response. Therefore, appropriate functional studies or other 7. Chen Z, Ji Z, Ngiow SF, Manne S, Cai Z, Huang AC, et  al. TCF-1-
immunologic assays may be necessary to identify additional centered transcriptional network drives an effector versus exhausted
biomarkers that should be included in the bioinformatic CD8 T cell-fate decision. Immunity 2019;51:840–55.
analyses for a more accurate definition of a given immune cell 8. Hudson WH, Gensheimer J, Hashimoto M, Wieland A, Valanparam-
state. Careful attention to these types of details will be neces- bil RM, Li P, et al. Proliferating transitory T cells with an effector-like
transcriptional signature emerge from PD-1(+) stem-like CD8(+) T
sary for advancing the field of ICT.
cells during chronic infection. Immunity 2019;51:1043–58.
9. McLane LM, Abdel-Hakeem MS, Wherry EJ. CD8 T cell exhaus-
CONCLUSIONS tion during chronic viral infection and cancer. Annu Rev Immunol
2019;37:457–95.
Over the next decade, the field of cancer immunotherapy 10. Alegre ML, Noel PJ, Eisfelder BJ, Chuang E, Clark MR, Reiner SL,
will need to grow vertically, to gain a deeper understanding et  al. Regulation of surface and intracellular expression of CTLA4
of different immune cell subsets and pathways that regulate on mouse T cells. J Immunol 1996;157:4762–70.
11. Takahashi T, Tagami T, Yamazaki S, Uede T, Shimizu J, Sakaguchi N,
their responses, as well as horizontally, to integrate with other
et  al. Immunologic self-tolerance maintained by CD25(+)CD4(+)
fields, including cancer biology, epigenetics, computational regulatory T cells constitutively expressing cytotoxic T lymphocyte-
biology, and many others. ICT will also need to expand across associated antigen 4. J Exp Med 2000;192:303–10.
all of oncology for the development of combination strategies, 12. Krummel MF, Allison JP. CD28 and CTLA-4 have opposing effects on
including combinations with surgery, radiotherapy, chemo- the response of T cells to stimulation. J Exp Med 1995;182:459–65.
therapy, targeted therapy, and other immunotherapy agents 13. Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clini-
cal application. Int Immunol 2007;19:813–24.
such as novel immune checkpoints and chimeric antigen
14. Inman BA, Sebo TJ, Frigola X, Dong H, Bergstralh EJ, Frank I, et al.
receptor T cells. As the field continues to grow, it will need to PD-L1 (B7-H1) expression by urothelial carcinoma of the bladder
remain grounded in the strength of basic immunology while and BCG-induced granulomata: associations with localized stage
it fosters close collaborations with other scientific disciplines. progression. Cancer 2007;109:1499–505.
The future of ICT is promising, with ample opportunity to 15. Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamagu-
further improve outcomes for patients with cancer. chi K, et al. Programmed cell death 1 ligand 1 and tumor-infiltrating
CD8+ T lymphocytes are prognostic factors of human ovarian can-
Authors’ Disclosures cer. Proc Natl Acad Sci U S A 2007;104:3360–5.
16. Im SJ, Hashimoto M, Gerner MY, Lee J, Kissick HT, Burger MC,
P. Sharma reports personal fees from Jounce Therapeutics, Onc- et al. Defining CD8+ T cells that provide the proliferative burst after
olytics, Earli, Glympse Bio, Lava Therapeutics, Lytix, Phenomics, PD-1 therapy. Nature 2016;537:417–21.
Codiak, Constellation, Achelois, Dragonfly, BioAtla, Infinity Pharma, 17. Carthon BC, Wolchok JD, Yuan J, Kamat A, Ng Tang DS, Sun J, et al.
Hummingbird, and Polaris outside the submitted work; in addition, Preoperative CTLA-4 blockade: tolerability and immune monitor-
P. Sharma has a patent for ARID1A plus CXCL13 as combinatorial ing in the setting of a presurgical clinical trial. Clin Cancer Res
biomarker pending and a patent for combination of anti-CTLA4 2010;16:2861–71.
plus agonist targeting of ICOS for improved antitumor immune 18. Liakou CI, Kamat A, Tang DN, Chen H, Sun J, Troncoso P, et  al.
responses licensed to Jounce Therapeutics. S.K. Subudhi reports per- CTLA-4 blockade increases IFNgamma-producing CD4+ICOShi
sonal fees from Dendreon, Kahr Medical, Ltd., Cancer Expert Now, cells to shift the ratio of effector to regulatory T cells in cancer
Inc., Bayer Healthcare Pharmaceuticals, Apricity Heatlh, LLC, Dava patients. Proc Natl Acad Sci U S A 2008;105:14987–92.

April 2021 CANCER DISCOVERY 10TH ANNIVERSARY ISSUE | 851

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
REVIEW Sharma et al.

19. Chen H, Liakou CI, Kamat A, Pettaway C, Ward JF, Tang DN, 39. Seymour L, Bogaerts J, Perrone A, Ford R, Schwartz LH, Mandrekar S,
et  al. Anti-CTLA-4 therapy results in higher CD4 +ICOS hi T cell et al. iRECIST: guidelines for response criteria for use in trials test-
frequency and IFNγ levels in both nonmalignant and malignant ing immunotherapeutics. Lancet Oncol 2017;18:e143–52.
prostate tissues. Proc Natl Acad Sci U S A 2009;106:2729–34. 40. Pignon JC, Jegede O, Shukla SA, Braun DA, Horak CE, Wind-Rotolo M,
20. Ng Tang D, Shen Y, Sun J, Wen S, Wolchok JD, Yuan J, et al. Increased et al. irRECIST for the evaluation of candidate biomarkers of response
frequency of ICOS+ CD4 T cells as a pharmacodynamic biomarker to nivolumab in metastatic clear cell renal cell carcinoma: analysis of
for anti-CTLA-4 therapy. Cancer Immunol Res 2013;1:229–34. a phase II prospective clinical trial. Clin Cancer Res 2019;25:2174–84.
21. Gao J, Navai N, Alhalabi O, Siefker-Radtke A, Campbell MT, Tidwell 41. Ayoub M, Eleneen Y, Colen RR. Cancer imaging in immunotherapy.
RS, et al. Neoadjuvant PD-L1 plus CTLA-4 blockade in patients with In: Naing A, Hajjar J, editors. Immunotherapy. Cham: Springer
cisplatin-ineligible operable high-risk urothelial carcinoma. Nat International Publishing; 2020. p. 309–24.
Med 2020;26:1845–51. 42. Schadendorf D, Hodi FS, Robert C, Weber JS, Margolin K, Hamid O,
22. Quezada SA, Peggs KS, Curran MA, Allison JP. CTLA4 blockade and et al. Pooled analysis of long-term survival data from phase II and
GM-CSF combination immunotherapy alters the intratumor balance phase III trials of ipilimumab in unresectable or metastatic mela-
of effector and regulatory T cells. J Clin Invest 2006;116:1935–45. noma. J Clin Oncol 2015;33:1889–94.
23. Peggs KS, Quezada SA, Chambers CA, Korman AJ, Allison JP. Block- 43. Perets R, Bar J, Rasco DW, Ahn M-J, Yoh K, Kim D-W, et al. Safety
ade of CTLA-4 on both effector and regulatory T cell compartments and efficacy of quavonlimab, a novel anti-CTLA-4 antibody (MK-
contributes to the antitumor activity of anti-CTLA-4 antibodies. 1308), in combination with pembrolizumab in first-line advanced
J Exp Med 2009;206:1717–25. non-small-cell lung cancer. Ann Oncol 2020;S0923–7534:43175–8.
24. Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, 44. Korman AJ, Engelhardt J, Loffredo J, Valle J, Akter R, Vuyyuru R,
Fehervari Z, et  al. CTLA-4 control over Foxp3+ regulatory T cell et  al. Next-generation anti-CTLA-4 antibodies [abstract]. In: Pro-
function. Science 2008;322:271–5. ceedings of the American Association for Cancer Research Annual
25. Read S, Greenwald R, Izcue A, Robinson N, Mandelbrot D, Fran- Meeting 2017; 2017 Apr 1–5; Washington, DC. Philadelphia (PA):
cisco L, et al. Blockade of CTLA-4 on CD4+CD25+ regulatory T cells AACR; 2017. Abstract nr SY09-01.
abrogates their function in vivo. J Immunol 2006;177:4376–83. 45. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al.
26. Romano E, Kusio-Kobialka M, Foukas PG, Baumgaertner P, Meyer Pembrolizumab versus ipilimumab in advanced melanoma. N Engl
C, Ballabeni P, et al. Ipilimumab-dependent cell-mediated cytotoxic- J Med 2015;372:2521–32.
ity of regulatory T cells ex vivo by nonclassical monocytes in mela- 46. Weber JS, D’Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, et al.
noma patients. Proc Natl Acad Sci U S A 2015;112:6140–5. Nivolumab versus chemotherapy in patients with advanced mela-
27. Sharma A, Subudhi SK, Blando J, Scutti J, Vence L, Wargo J, et al. noma who progressed after anti-CTLA-4 treatment (CheckMate 037):
Anti-CTLA-4 immunotherapy does not deplete Foxp3 þ regulatory a randomised, controlled, open-label, phase 3 trial. Lancet Oncol
T cells (Tregs) in human cancers. Clin Cancer Res 2019;25:1233–8. 2015;16:375–84.
28. Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 47. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD,
combination blockade expands infiltrating T cells and reduces et al. PD-1 blockade in tumors with mismatch-repair deficiency. N
regulatory T and myeloid cells within B16 melanoma tumors. Proc Engl J Med 2015;372:2509–20.
Natl Acad Sci U S A 2010;107:4275–80. 48. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK,
29. Wei SC, Levine JH, Cogdill AP, Zhao Y, Anang NAAS, Andrews MC, et al. Mismatch repair deficiency predicts response of solid tumors
et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti- to PD-1 blockade. Science 2017;357:409–13.
PD-1 checkpoint blockade. Cell 2017;170:1120–33. 49. Marabelle A, Fakih M, Lopez J, Shah M, Shapira-Frommer R,
30. Wei SC, Sharma R, Anang N-AAS, Levine JH, Zhao Y, Mancuso JJ, Nakagawa K, et  al. Association of tumour mutational burden with
et  al. Negative co-stimulation constrains T cell differentiation by outcomes in patients with advanced solid tumours treated with pem-
imposing boundaries on possible cell states. Immunity 2019;50: brolizumab: prospective biomarker analysis of the multicohort, open-
1084–98. label, phase 2 KEYNOTE-158 study. Lancet Oncol 2020;21:1353–65.
31. Berger R, Rotem-Yehudar R, Slama G, Landes S, Kneller A, Leiba M, 50. Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R,
et al. Phase I safety and pharmacokinetic study of CT-011, a human- et al. Durvalumab after chemoradiotherapy in stage III non–small-
ized antibody interacting with PD-1, in patients with advanced cell lung cancer. N Engl J Med 2017;377:1919–29.
hematologic malignancies. Clin Cancer Res 2008;14:3044–51. 51. Horn L, Mansfield AS, Szczȩsna A, Havel L, Krzakowski M, Hoch-
32. Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman mair MJ, et al. First-line atezolizumab plus chemotherapy in exten-
WH, et  al. Phase I study of single-agent anti-programmed death-1 sive-stage small-cell lung cancer. N Engl J Med 2018;379:2220–9.
(MDX-1106) in refractory solid tumors: safety, clinical activity, phar- 52. Paz-Ares L, Dvorkin M, Chen Y, Reinmuth N, Hotta K, Trukhin D,
macodynamics, and immunologic correlates. J Clin Oncol 2010; et al. Durvalumab plus platinum–etoposide versus platinum–etopo-
28:3167–75. side in first-line treatment of extensive-stage small-cell lung cancer
33. Brahmer JR, Tykodi SS, Chow LQM, Hwu W-J, Topalian SL, Hwu (CASPIAN): a randomised, controlled, open-label, phase 3 trial.
P, et al. Safety and activity of anti-PD-L1 antibody in patients with Lancet 2019;394:1929–39.
advanced cancer. N Engl J Med 2012;366:2455–65. 53. Powles T, Park SH, Voog E, Caserta C, Valderrama BP, Gurney H,
34. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, et  al. Avelumab maintenance therapy for advanced or metastatic
McDermott DF, et al. Safety, activity, and immune correlates of anti- urothelial carcinoma. N Engl J Med 2020;383:1218–30.
PD-1 antibody in cancer. N Engl J Med 2012;366:2443–54. 54. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD,
35. Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint et  al. Combined nivolumab and ipilimumab or monotherapy in
blockade therapy for cancer: an overview of FDA-approved immune untreated melanoma. N Engl J Med 2015;373:23–34.
checkpoint inhibitors. Int Immunopharmacol 2018;62:29–39. 55. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao
36. Vaddepally RK, Kharel P, Pandey R, Garje R, Chandra AB. Review CD, et  al. Five-year survival with combined nivolumab and ipili-
of indications of FDA-approved immune checkpoint inhibitors per mumab in advanced melanoma. N Engl J Med 2019;381:1535–46.
NCCN guidelines with the level of evidence. Cancers 2020;12:738. 56. Yau T, Kang Y-K, Kim T-Y, El-Khoueiry AB, Santoro A, Sangro B, et al. Effi-
37. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, cacy and safety of nivolumab plus ipilimumab in patients with advanced
et al. Improved survival with ipilimumab in patients with metastatic hepatocellular carcinoma previously treated with sorafenib: the Check-
melanoma. N Engl J Med 2010;363:711–23. Mate 040 randomized clinical trial. JAMA Oncol 2020;6:e204564.
38. Robert C, Thomas L, Bondarenko I, O’Day S, Weber J, Garbe C, et al. 57. Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar
Ipilimumab plus dacarbazine for previously untreated metastatic B, Choueiri TK, et al. Nivolumab plus ipilimumab versus sunitinib
melanoma. N Engl J Med 2011;364:2517–26. in advanced renal-cell carcinoma. N Engl J Med 2018;378:1277–90.

852 | CANCER DISCOVERY 10TH ANNIVERSARY ISSUE April 2021 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
The Next Decade of Immune Checkpoint Therapy REVIEW

58. Motzer RJ, Escudier B, McDermott DF, Arén Frontera O, Melichar B, 76. Gutzmer R, Stroyakovskiy D, Gogas H, Robert C, Lewis K, Prot-
Powles T, et al. Survival outcomes and independent response assess- senko S, et  al. Atezolizumab, vemurafenib, and cobimetinib as
ment with nivolumab plus ipilimumab versus sunitinib in patients first-line treatment for unresectable advanced BRAFV600 mutation-
with advanced renal cell carcinoma: 42-month follow-up of a rand- positive melanoma (IMspire150): primary analysis of the ran-
omized phase 3 clinical trial. J Immunother Cancer 2020;8:e000891. domised, double-blind, placebo-controlled, phase 3 trial. Lancet 2020;
59. Hellmann MD, Paz-Ares L, Bernabe Caro R, Zurawski B, Kim S-W, 395:1835–44.
Carcereny Costa E, et al. Nivolumab plus ipilimumab in advanced 77. Brahmer JR, Lacchetti C, Schneider BJ, Atkins MB, Brassil KJ,
non–small-cell lung cancer. N Engl J Med 2019;381:2020–31. Caterino JM, et al. Management of immune-related adverse events
60. Scherpereel A, Mazieres J, Greillier L, Lantuejoul S, Dô P, Bylicki O, in patients treated with immune checkpoint inhibitor therapy:
et  al. Nivolumab or nivolumab plus ipilimumab in patients with American Society of Clinical Oncology clinical practice guideline.
relapsed malignant pleural mesothelioma (IFCT-1501 MAPS2): a J Clin Oncol 2018;36:1714–68.
multicentre, open-label, randomised, non-comparative, phase 2 78. Abu-Sbeih H, Ali FS, Wang X, Mallepally N, Chen E, Altan M, et al.
trial. Lancet Oncol 2019;20:239–53. Early introduction of selective immunosuppressive therapy associ-
61. Overman MJ, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, ated with favorable clinical outcomes in patients with immune
et  al. Durable clinical benefit with nivolumab plus ipilimumab checkpoint inhibitor-induced colitis. J Immunother Cancer 2019;
in DNA mismatch repair-deficient/microsatellite instability-high 7:1–11.
metastatic colorectal cancer. J Clin Oncol 2018;36:773–9. 79. Subudhi SK, Aparicio A, Gao J, Zurita AJ, Araujo JC, Logothetis CJ,
62. Leonetti A, Wever B, Mazzaschi G, Assaraf YG, Rolfo C, Quaini F, et  al. Clonal expansion of CD8 T cells in the systemic circulation
et al. Molecular basis and rationale for combining immune check- precedes development of ipilimumab-induced toxicities. Proc Natl
point inhibitors with chemotherapy in non-small cell lung cancer. Acad Sci U S A 2016;113:11919–24.
Drug Resist Updat 2019;46:100644. 80. Tahir SA, Gao J, Miura Y, Blando J, Tidwell RSS, Zhao H, et al. Auto-
63. Gandhi L, Rodríguez-Abreu D, Gadgeel S, Esteban E, Felip E, De immune antibodies correlate with immune checkpoint therapy-
Angelis F, et  al. Pembrolizumab plus chemotherapy in metastatic induced toxicities. Proc Natl Acad Sci U S A 2019;116:22246–51.
non-small-cell lung cancer. N Engl J Med 2018;378:2078–92. 81. Campbell MT, Zhang T, Chin L, Warner AB, Mathew M. Apric-
64. Paz-Ares L, Luft A, Vicente D, Tafreshi A, Gümüş M, Mazières J, et al. ityRx companion digital therapeutic for evidence-based mitiga-
Pembrolizumab plus chemotherapy for squamous non-small-cell tion and phenotype-linked molecular characterization of irAEs in
lung cancer. N Engl J Med 2018;379:2040–51. patients receiving immune checkpoint therapy (ICT). J Clin Oncol
65. Socinski MA, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, 2020;38:TPS2089.
Nogami N, et al. Atezolizumab for first-line treatment of metastatic 82. Maher VE, Fernandes LL, Weinstock C, Tang S, Agarwal S, Brave M,
nonsquamous NSCLC. N Engl J Med 2018;378:2288–301. et  al. Analysis of the association between adverse events and out-
66. West H, McCleod M, Hussein M, Morabito A, Rittmeyer A, Conter come in patients receiving a programmed death protein 1 or pro-
HJ, et al. Atezolizumab in combination with carboplatin plus nab- grammed death ligand 1 antibody. J Clin Oncol 2019;37:2730–7.
paclitaxel chemotherapy compared with chemotherapy alone as 83. Indini A, Di Guardo L, Cimminiello C, Prisciandaro M, Randon G,
first-line treatment for metastatic non-squamous non-small-cell De Braud F, et  al. Immune-related adverse events correlate with
lung cancer (IMpower130): a multicentre, randomised, open-label, improved survival in patients undergoing anti-PD1 immunotherapy
phase 3 tria. Lancet Oncol 2019;20:924–37. for metastatic melanoma. J Cancer Res Clin Oncol 2019;145:511–21.
67. Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH, Iwata H, 84. Sato K, Akamatsu H, Murakami E, Sasaki S, Kanai K, Hayata A, et al.
et  al. Atezolizumab and nab-paclitaxel in advanced triple-negative Correlation between immune-related adverse events and efficacy in
breast cancer. N Engl J Med 2018;379:2108–21. non-small cell lung cancer treated with nivolumab. Lung Cancer
68. Burtness B, Harrington KJ, Greil R, Soulières D, Tahara M, de Castro 2018;115:71–4.
G, et al. Pembrolizumab alone or with chemotherapy versus cetuxi- 85. Mian I, Yang M, Zhao H, Shah M, Diab A, Shannon V, et  al.
mab with chemotherapy for recurrent or metastatic squamous cell Immune-related adverse events and survival in elderly patients with
carcinoma of the head and neck (KEYNOTE-048): a randomised, melanoma treated with ipilimumab. J Clin Oncol 2016;34:3047.
open-label, phase 3 study. Lancet 2019;394:1915–28. 86. Owen DH, Wei L, Bertino EM, Edd T, Villalona-Calero MA, He K,
69. Makker V, Rasco D, Vogelzang NJ, Brose MS, Cohn AL, Mier J, et al. et  al. Incidence, risk factors, and effect on survival of immune-
Lenvatinib plus pembrolizumab in patients with advanced endome- related adverse events in patients with non-small-cell lung cancer.
trial cancer: an interim analysis of a multicentre, open-label, single- Clin Lung Cancer 2018;19:e893–900.
arm, phase 2 trial. Lancet Oncol 2019;20:711–8. 87. Horvat TZ, Adel NG, Dang T-O, Momtaz P, Postow MA, Callahan
70. Makker V, Taylor MH, Aghajanian C, Oaknin A, Mier J, Cohn AL, MK, et al. Immune-related adverse events, need for systemic immu-
et  al. Lenvatinib plus pembrolizumab in patients with advanced nosuppression, and effects on survival and time to treatment failure
endometrial cancer. J Clin Oncol 2020;38:2981–92. in patients with melanoma treated with ipilimumab at Memorial
71. Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, et al. Sloan Kettering Cancer Center. J Clin Oncol 2015;33:3193–8.
Pembrolizumab plus axitinib versus sunitinib for advanced renal- 88. Ricciuti B, Genova C, De Giglio A, Bassanelli M, Dal Bello MG,
cell carcinoma. N Engl J Med 2019;380:1116–27. Metro G, et al. Impact of immune-related adverse events on survival
72. Plimack ER, Rini BI, Stus V, Gafanov R, Waddell T, Nosov D, et al. in patients with advanced non-small cell lung cancer treated with
Pembrolizumab plus axitinib versus sunitinib as first-line ther- nivolumab: long-term outcomes from a multi-institutional analysis.
apy for advanced renal cell carcinoma (RCC): updated analysis of J Cancer Res Clin Oncol 2019;145:479–85.
KEYNOTE-426. J Clin Oncol 2020;38:5001. 89. Xing P, Zhang F, Wang G, Xu Y, Li C, Wang S, et al. Incidence rates of
73. Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, immune-related adverse events and their correlation with response
et al. Avelumab plus axitinib versus sunitinib for advanced renal-cell in advanced solid tumours treated with NIVO or NIVO+IPI: a sys-
carcinoma. N Engl J Med 2019;380:1103–15. tematic review and meta-analysis. J Immunother Cancer 2019;7:341.
74. Choueiri TK, Powles T, Burotto M, Bourlon MT, Zurawski B, 90. Das S, Johnson DB. Immune-related adverse events and anti-tumor
Oyervides Juárez VM, et al. 696O_PR Nivolumab + cabozantinib vs efficacy of immune checkpoint inhibitors. J Immunother Cancer
sunitinib in first-line treatment for advanced renal cell carcinoma: 2019;7:306.
first results from the randomized phase III CheckMate 9ER trial. 91. Riudavets M, Mosquera J, Garcia-Campelo R, Serra J, Anguera G,
Ann Oncol 2020;31:S1159. Gallardo P, et  al. Immune-related adverse events and corticoster-
75. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim T-Y, et  al. oid use for cancer-related symptoms are associated with efficacy
Atezolizumab plus bevacizumab in unresectable hepatocellular car- in patients with non-small cell lung cancer receiving anti-PD-(L)1
cinoma. N Engl J Med 2020;382:1894–905. blockade agents. Front Oncol 2020;10:1677.

April 2021 CANCER DISCOVERY 10TH ANNIVERSARY ISSUE | 853

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
REVIEW Sharma et al.

92. Rogado J, Sánchez-Torres JM, Romero-Laorden N, Ballesteros AI, prostate cancer: preliminary analysis of patients in the CheckMate
Pacheco-Barcia V, Ramos-Leví A, et  al. Immune-related adverse 650 trial. Cancer Cell 2020;38:489–99.
events predict the therapeutic efficacy of anti-PD-1 antibodies in 110. Jiao S, Subudhi SK, Aparicio A, Ge Z, Guan B, Miura Y, et al. Differences
cancer patients. Eur J Cancer 2019;109:21–7. in tumor microenvironment dictate T helper lineage polarization and
93. Toi Y, Sugawara S, Kawashima Y, Aiba T, Kawana S, Saito R, et al. response to immune checkpoint therapy. Cell 2019;179:1177–90.
Association of immune-related adverse events with clinical benefit 111. Lee JC, Mehdizadeh S, Smith J, Young A, Mufazalov IA, Mowery CT,
in patients with advanced non-small-cell lung cancer treated with et al. Regulatory T cell control of systemic immunity and immuno-
nivolumab. Oncologist 2018;23:1358–65. therapy response in liver metastasis. Sci Immunol 2020;5:eaba0759.
94. Okada N, Kawazoe H, Takechi K, Matsudate Y, Utsunomiya R, Zamami Y, 112. Goswami S, Walle T, Cornish AE, Basu S, Anandhan S, Fernandez
et al. Association between immune-related adverse events and clinical I, et  al. Immune profiling of human tumors identifies CD73 as a
efficacy in patients with melanoma treated with nivolumab: a multi- combinatorial target in glioblastoma. Nat Med 2020;26:39–46.
center retrospective study. Clin Ther 2019;41:59–67. 113. Najafi M, Hashemi Goradel N, Farhood B, Salehi E, Nashtaei MS,
95. Elias R, Yan F, Singla N, Levonyack N, Formella J, Christie A, et al. Khanlarkhani N, et al. Macrophage polarity in cancer: a review. J Cell
Immune-related adverse events are associated with improved out- Biochem 2019;120:2756–65.
comes in ICI-treated renal cell carcinoma patients. J Clin Oncol 2019; 114. Asare PF, Roscioli E, Hurtado PR, Tran HB, Mah CY, Hodge S. LC3-
37:645. associated phagocytosis (LAP): a potentially influential mediator of
96. Grangeon M, Tomasini P, Chaleat S, Jeanson A, Souquet-Bressand M, efferocytosis-related tumor progression and aggressiveness. Front
Khobta N, et al. Association between immune-related adverse events Oncol 2020;10:1298.
and efficacy of immune checkpoint inhibitors in non-small-cell 115. Flavahan WA, Gaskell E, Bernstein BE. Epigenetic plasticity and the
lung cancer. Clin Lung Cancer 2019;20:201–7. hallmarks of cancer. Science 2017;357:eaal2380.
97. Wei SC, Meijers WC, Axelrod ML, Anang N-AAS, Screever EM, 116. Goswami S, Apostolou I, Zhang J, Skepner J, Anandhan S, Zhang X,
Wescott EC, et  al. A genetic mouse model recapitulates immune et al. Modulation of EZH2 expression in T cells improves efficacy of
checkpoint inhibitor-associated myocarditis and supports a mecha- anti-CTLA-4 therapy. J Clin Invest 2018;128:3813–8.
nism-based therapeutic intervention. Cancer Discov 2021;11:1–12. 117. Suvà ML, Riggi N, Bernstein BE. Epigenetic reprogramming in can-
98. Salem J-E, Allenbach Y, Vozy A, Brechot N, Johnson DB, Moslehi JJ, cer. Science 2013;339:1567–70.
et al. Abatacept for severe immune checkpoint inhibitor–associated 118. Kim KH, Roberts CWM. Targeting EZH2 in cancer. Nat Med 2016;
myocarditis. N Engl J Med 2019;380:2377–9. 22:128–34.
99. Luoma AM, Suo S, Williams HL, Sharova T, Sullivan K, Manos M, 119. DuPage M, Chopra G, Quiros J, Rosenthal WL, Morar MM, Holo-
et al. Molecular pathways of colon inflammation induced by cancer han D, et  al. The chromatin-modifying enzyme Ezh2 is critical
immunotherapy. Cell 2020;182:655–71. for the maintenance of regulatory T cell identity after activation.
100. Kwon ED, Drake CG, Scher HI, Fizazi K, Bossi A, van den Eertwegh Immunity 2015;42:227–38.
AJM, et al. Ipilimumab versus placebo after radiotherapy in patients 120. Villanueva L, Álvarez-Errico D, Esteller M. The contribution of epige-
with metastatic castration-resistant prostate cancer that had pro- netics to cancer immunotherapy. Trends Immunol 2020;41:676–91.
gressed after docetaxel chemotherapy (CA184-043): a multicentre, ran- 121. Sharma P, Allison JP. Immune checkpoint targeting in cancer ther-
domised, double-blind, phase 3 trial. Lancet Oncol 2014;15:700–12. apy: toward combination strategies with curative potential. Cell
101. Beer TM, Kwon ED, Drake CG, Fizazi K, Logothetis C, Gravis G, 2015;161:205–14.
et al. Randomized, double-blind, phase III trial of ipilimumab ver- 122. Hamid O, Schmidt H, Nissan A, Ridolfi L, Aamdal S, Hansson J,
sus placebo in asymptomatic or minimally symptomatic patients et al. A prospective phase II trial exploring the association between
with metastatic chemotherapy-naive castration-resistant prostate tumor microenvironment biomarkers and clinical activity of ipili-
cancer. J Clin Oncol 2017;35:40–7. mumab in advanced melanoma. J Transl Med 2011;9:204.
102. Antonarakis ES, Piulats JM, Gross-Goupil M, Goh J, Ojamaa K, 123. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin sig-
Hoimes CJ, et  al. Pembrolizumab for treatment-refractory meta- nalling prevents anti-tumour immunity. Nature 2015;523:231–5.
static castration-resistant prostate cancer: multicohort, open-label 124. Peng W, Chen JQ, Liu C, Malu S, Creasy C, Tetzlaff MT, et al. Loss
phase II KEYNOTE-199 study. J Clin Oncol 2020;38:395–405. of PTEN promotes resistance to T cell-mediated immunotherapy.
103. Sweeney CJ, Gillessen S, Rathkopf D, Matsubara N, Drake C, Fizazi Cancer Discov 2016;6:202–16.
K, et al. Abstract CT014: IMbassador250: a phase III trial comparing 125. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard
atezolizumab with enzalutamide vs enzalutamide alone in patients A, et  al. Genetic basis for clinical response to CTLA-4 blockade in
with metastatic castration-resistant prostate cancer (mCRPC). melanoma. N Engl J Med 2014;371:2189–99.
Cancer Res 2020;CT014 LP–CT014. 126. Hellmann MD, Nathanson T, Rizvi H, Creelan BC, Sanchez-Vega F,
104. Antonarakis ES, Goh JC, Gross-Goupil M, Vaishampayan UN, Piu- Ahuja A, et al. Genomic features of response to combination immu-
lats JM, De Wit R, et al. Pembrolizumab for metastatic castration- notherapy in patients with advanced non-small-cell lung cancer.
resistant prostate cancer (mCRPC) previously treated with docetaxel: Cancer Cell 2018;33:843–52.
updated analysis of KEYNOTE-199. J Clin Oncol 2019;37:216. 127. Ganesan S, Mehnert J. Biomarkers for response to immune check-
105. Gao J, Ward JF, Pettaway CA, Shi LZ, Subudhi SK, Vence LM, et al. VISTA point blockade. Annu Rev Cancer Biol 2020;4:331–51.
is an inhibitory immune checkpoint that is increased after ipilimumab 128. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and
therapy in patients with prostate cancer. Nat Med 2017;23:551–5. response rate to PD-1 inhibition. N Engl J Med 2017;377:2500–1.
106. Bishop JL, Sio A, Angeles A, Roberts ME, Azad AA, Chi KN, et  al. 129. Braun DA, Bakouny Z, Hirsch L, Flippot R, Van Allen EM, Wu CJ,
PD-L1 is highly expressed in enzalutamide resistant prostate cancer. et al. Beyond conventional immune-checkpoint inhibition — novel
Oncotarget 2015;6:234–42. immunotherapies for renal cell carcinoma. Nat Rev Clin Oncol 2021
107. Fizazi K, Drake CG, Beer TM, Kwon ED, Scher HI, Gerritsen WR, Jan 12 [Epub ahead of print].
et  al. Final analysis of the ipilimumab versus placebo following 130. Cristescu R, Mogg R, Ayers M, Albright A, Murphy E, Yearley J, et al.
radiotherapy phase III trial in postdocetaxel metastatic castration- Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-
resistant prostate cancer identifies an excess of long-term survivors. based immunotherapy. Science 2018;362:eaar3593.
Eur Urol 2020;78:822–30. 131. Subudhi SK, Vence L, Zhao H, Blando J, Yadav SS, Xiong Q, et al.
108. Lines JL, Pantazi E, Mak J, Sempere LF, Wang L, O’Connell S, et al. Neoantigen responses, immune correlates, and favorable outcomes
VISTA is an immune checkpoint molecule for human T cells. Can- after ipilimumab treatment of patients with prostate cancer. Sci
cer Res 2014;74:1924–32. Transl Med 2020;12:1–11.
109. Sharma P, Pachynski RK, Narayan V, Fléchon A, Gravis G, Galsky MD, 132. Braun DA, Hou Y, Bakouny Z, Ficial M, Sant’ Angelo M, Forman J,
et al. Nivolumab plus ipilimumab for metastatic castration-resistant et  al. Interplay of somatic alterations and immune infiltration

854 | CANCER DISCOVERY 10TH ANNIVERSARY ISSUE April 2021 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
The Next Decade of Immune Checkpoint Therapy REVIEW

modulates response to PD-1 blockade in advanced clear cell renal 151. Galon J, Fox BA, Bifulco CB, Masucci G, Rau T, Botti G, et al. Immu-
cell carcinoma. Nat Med 2020;26:909–18. noscore and immunoprofiling in cancer: an update from the mela-
133. Ott PA, Hu-Lieskovan S, Chmielowski B, Govindan R, Naing A, noma and immunotherapy bridge 2015. J Transl Med 2016;14:273.
Bhardwaj N, et al. A phase Ib trial of personalized neoantigen ther- 152. Bindea G, Mlecnik B, Angell HK, Galon J. The immune landscape of
apy plus anti-PD-1 in patients with advanced melanoma, non-small human tumors: implications for cancer immunotherapy. Oncoim-
cell lung cancer, or bladder cancer. Cell 2020;183:347–62. munology 2014;3:e27456.
134. Keskin DB, Anandappa AJ, Sun J, Tirosh I, Mathewson ND, Li S, 153. Sade-Feldman M, Yizhak K, Bjorgaard SL, Ray JP, de Boer CG,
et al. Neoantigen vaccine generates intratumoral T cell responses in Jenkins RW, et al. Defining T cell states associated with response to
phase Ib glioblastoma trial. Nature 2019;565:234–9. checkpoint immunotherapy in melanoma. Cell 2018;175:998–1013.
135. Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, et  al. An 154. Balatoni T, Mohos A, Papp E, Sebestyén T, Liszkay G, Oláh J, et al.
immunogenic personal neoantigen vaccine for patients with mela- Tumor-infiltrating immune cells as potential biomarkers predicting
noma. Nature 2017;547:217–21. response to treatment and survival in patients with metastatic mela-
136. Stewart RA, Pilie PG, Yap TA. Development of PARP and immune- noma receiving ipilimumab therapy. Cancer Immunol Immunother
checkpoint inhibitor combinations. Cancer Res 2018;78:6717–25. 2018;67:141–51.
137. Sen T, Rodriguez BL, Chen L, Della Corte CM, Morikawa N, Fuji- 155. Luke JJ, Bao R, Sweis RF, Spranger S, Gajewski TF. WNT/β-catenin
moto J, et al. Targeting DNA damage response promotes antitumor pathway activation correlates with immune exclusion across human
immunity through STING-mediated T-cell activation in small cell cancers. Clin Cancer Res 2019;25:3074–83.
lung cancer. Cancer Discov 2019;9:646–61. 156. Trujillo JA, Luke JJ, Zha Y, Segal JP, Ritterhouse LL, Spranger S, et al.
138. Parkes EE, Walker SM, Taggart LE, McCabe N, Knight LA, Wilkin- Secondary resistance to immunotherapy associated with β-catenin
son R, et al. Activation of STING-dependent innate immune signal- pathway activation or PTEN loss in metastatic melanoma. J Immu-
ing by s-phase-specific DNA damage in breast cancer. J Natl Cancer nother Cancer 2019;7:295.
Inst 2017;109:1–10. 157. Dighe AS, Richards E, Old LJ, Schreiber RD. Enhanced in vivo
139. Härtlova A, Erttmann SF, Raffi FA, Schmalz AM, Resch U, Anugula growth and resistance to rejection of tumor cells expressing domi-
S, et  al. DNA damage primes the type I interferon system via the nant negative IFN gamma receptors. Immunity 1994;1:447–56.
cytosolic DNA sensor STING to promote anti-microbial innate 158. Kaplan DH, Shankaran V, Dighe AS, Stockert E, Aguet M, Old LJ,
immunity. Immunity 2015;42:332–43. et  al. Demonstration of an interferon gamma-dependent tumor
140. Teo MY, Seier K, Ostrovnaya I, Regazzi AM, Kania BE, Moran MM, surveillance system in immunocompetent mice. Proc Natl Acad Sci
et  al. Alterations in DNA damage response and repair genes as U S A 1998;95:7556–61.
potential marker of clinical benefit from PD-1/PD-L1 blockade in 159. Gao J, Shi LZ, Zhao H, Chen J, Xiong L, He Q, et al. Loss of IFNγ
advanced urothelial cancers. J Clin Oncol 2018;36:1685–94. pathway genes in tumor cells as a mechanism of resistance to anti-
141. Samstein RM, Krishna C, Ma X, Pei X, Lee K-W, Makarov V, et  al. CTLA-4 therapy. Cell 2016;167:397–404.
Mutations in BRCA1 and BRCA2 differentially affect the tumor 160. Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H,

microenvironment and response to checkpoint blockade immuno- Hugo W, Hu-Lieskovan S, et  al. Mutations associated with
therapy. Nat Cancer 2020;1:1188–203. acquired resistance to PD-1 blockade in melanoma. N Engl J Med
142. Miao D, Margolis CA, Vokes NI, Liu D, Taylor-Weiner A, Wanko- 2016;375:819–29.
wicz SM, et  al. Genomic correlates of response to immune check- 161. Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman
point blockade in microsatellite-stable solid tumors. Nat Genet DR, et al. IFNγ–related mRNA profile predicts clinical response to
2018;50:1271–81. PD-1 blockade. J Clin Invest 2017;127:2930–40.
143. Rosenberg JE, Hoffman-Censits J, Powles T, Van Der Heijden MS, 162. Grasso CS, Tsoi J, Onyshchenko M, Abril-Rodriguez G, Ross-
Balar AV, Necchi A, et  al. Atezolizumab in patients with locally Macdonald P, Wind-Rotolo M, et al. Conserved interferon-γ signal-
advanced and metastatic urothelial carcinoma who have progressed ing drives clinical response to immune checkpoint blockade therapy
following treatment with platinum-based chemotherapy: a single- in melanoma. Cancer Cell 2020;38:500–15.
arm, multicentre, phase 2 trial. Lancet 2016;387:1909–20. 163. Petitprez F, de Reyniès A, Keung EZ, Chen TW-W, Sun C-M, Calde-
144. Balar AV, Galsky MD, Rosenberg JE, Powles T, Petrylak DP, Bellmunt raro J, et al. B cells are associated with survival and immunotherapy
J, et  al. Atezolizumab as first-line treatment in cisplatin-ineligible response in sarcoma. Nature 2020;577:556–60.
patients with locally advanced and metastatic urothelial carcinoma: 164. Helmink BA, Reddy SM, Gao J, Zhang S, Basar R, Thakur R, et al.
a single-arm, multicentre, phase 2 trial. Lancet 2017;389:67–76. B cells and tertiary lymphoid structures promote immunotherapy
145. Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, response. Nature 2020;577:549–55.
Toy ST, et  al. Use of tumor-infiltrating lymphocytes and interleu- 165. Koti M, Xu AS, Ren KYM, Visram K, Ren R, Berman DM, et al. Ter-
kin-2 in the immunotherapy of patients with metastatic melanoma. tiary lymphoid structures associate with tumour stage in urothelial
A preliminary report. N Engl J Med 1988;319:1676–80. bladder cancer. Bladder Cancer 2017;3:259–67.
146. Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, 166. Germain C, Gnjatic S, Tamzalit F, Knockaert S, Remark R, Goc J,
Regnani G, et  al. Intratumoral T cells, recurrence, and survival in et  al. Presence of B cells in tertiary lymphoid structures is associ-
epithelial ovarian cancer. N Engl J Med 2003;348:203–13. ated with a protective immunity in patients with lung cancer. Am J
147. Sharma P, Shen Y, Wen S, Yamada S, Jungbluth AA, Gnjatic S, et al. Respir Crit Care Med 2014;189:832–44.
CD8 tumor-infiltrating lymphocytes are predictive of survival in 167. Thommen DS, Koelzer VH, Herzig P, Roller A, Trefny M, Dimeloe S,
muscle-invasive urothelial carcinoma. Proc Natl Acad Sci U S A et al. A transcriptionally and functionally distinct PD-1(+) CD8(+)
2007;104:3967–72. T cell pool with predictive potential in non-small-cell lung cancer
148. Hodi FS, Butler M, Oble DA, Seiden MV, Haluska FG, Kruse A, et al. treated with PD-1 blockade. Nat Med 2018;24:994–1004.
Immunologic and clinical effects of antibody blockade of cytotoxic 168. Sautès-Fridman C, Petitprez F, Calderaro J, Fridman WH. Tertiary
T lymphocyte-associated antigen 4 in previously vaccinated cancer lymphoid structures in the era of cancer immunotherapy. Nat Rev
patients. Proc Natl Acad Sci U S A 2008;105:3005–10. Cancer 2019;19:307–25.
149. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJM, Robert 169. Litchfield K, Reading JL, Puttick C, Thakkar K, Abbosh C, Bentham
L, et  al. PD-1 blockade induces responses by inhibiting adaptive R, et al. Meta-analysis of tumor- and T cell-intrinsic mechanisms of
immune resistance. Nature 2014;515:568–71. sensitization to checkpoint inhibition. Cell 2021;184:596–614.
150. Loi S, Drubay D, Adams S, Pruneri G, Francis PA, Lacroix-Triki M, 170. Goswami S, Chen Y, Anandhan S, Szabo PM, Basu S, Blando JM,
et al. Tumor-infiltrating lymphocytes and prognosis: a pooled indi- et al. ARID1A mutation plus CXCL13 expression act as combinato-
vidual patient analysis of early-stage triple-negative breast cancers. rial biomarkers to predict responses to immune checkpoint therapy
J Clin Oncol 2019;37:559–69. in mUCC. Sci Transl Med 2020;12:eabc4220.

April 2021 CANCER DISCOVERY 10TH ANNIVERSARY ISSUE | 855

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
REVIEW Sharma et al.

171. Fu T, He Q, Sharma P. The ICOS/ICOSL pathway is required for 187. Forde PM, Chaft JE, Felip E, Broderick S, Girard N, Awad MM,
optimal antitumor responses mediated by anti-CTLA-4 therapy. et  al. Checkmate 816: a phase 3, randomized, open-label trial of
Cancer Res 2011;71:5445–54. nivolumab plus ipilimumab vs platinum-doublet chemotherapy as
172. Fan X, Quezada SA, Sepulveda MA, Sharma P, Allison JP. Engage- neoadjuvant treatment for early-stage NSCLC. J Clin Oncol 2017;
ment of the ICOS pathway markedly enhances efficacy of CTLA-4 35:TPS8577.
blockade in cancer immunotherapy. J Exp Med 2014;211:715–25. 188. Sonpavde G, Necchi A, Gupta S, Steinberg GD, Gschwend JE, Van
173. Amaria RN, Reddy SM, Tawbi HA, Davies MA, Ross MI, Glitza IC, Der Heijden MS, et al. A phase 3 randomized study of neoadjuvant
et al. Neoadjuvant immune checkpoint blockade in high-risk resect- chemotherapy (NAC) alone or in combination with nivolumab
able melanoma. Nat Med 2018;24:1649–54. (NIVO) ± BMS-986205 in cisplatin-eligible muscle invasive bladder
174. Rozeman EA, Menzies AM, van Akkooi ACJ, Adhikari C, Bierman C, cancer (MIBC). J Clin Oncol 2019;37:TPS4587.
van de Wiel BA, et al. Identification of the optimal combination dos- 189. Eggermont AMM, Blank CU, Mandala M, Long GV, Atkinson V,
ing schedule of neoadjuvant ipilimumab plus nivolumab in macro- Dalle S, et al. Adjuvant pembrolizumab versus placebo in resected
scopic stage III melanoma (OpACIN-neo): a multicentre, phase 2, stage III melanoma. N Engl J Med 2018;378:1789–801.
randomised, controlled trial. Lancet Oncol 2019;20:948–60. 190. Eggermont AMM, Chiarion-Sileni V, Grob J-J, Dummer R, Wolchok
175. Nghiem P, Bhatia S, Lipson EJ, Sharfman WH, Kudchadkar RR, JD, Schmidt H, et  al. Adjuvant ipilimumab versus placebo after
Brohl AS, et  al. Durable tumor regression and overall survival in complete resection of high-risk stage III melanoma (EORTC 18071):
patients with advanced Merkel cell carcinoma receiving pembroli- a randomised, double-blind, phase 3 trial. Lancet Oncol 2015;
zumab as first-line therapy. J Clin Oncol 2019;37:693–702. 16:522–30.
176. Forde PM, Chaft JE, Smith KN, Anagnostou V, Cottrell TR, Hell- 191. Weber J, Mandala M, Del Vecchio M, Gogas HJ, Arance AM, Cowey CL,
mann MD, et  al. Neoadjuvant PD-1 blockade in resectable lung et al. Adjuvant nivolumab versus ipilimumab in resected stage III or
cancer. N Engl J Med 2018;378:1976–86. IV melanoma. N Engl J Med 2017;377:1824–35.
177. Chalabi M, Fanchi LF, Dijkstra KK, Van den Berg JG, Aalbers AG, 192. Tarhini AA, Lee SJ, Hodi FS, Rao UNM, Cohen GI, Hamid O, et al.
Sikorska K, et  al. Neoadjuvant immunotherapy leads to patho- Phase III study of adjuvant ipilimumab (3 or 10 mg/kg) versus high-
logical responses in MMR-proficient and MMR-deficient early-stage dose interferon alfa-2b for resected high-risk melanoma: North
colon cancers. Nat Med 2020;26:566–76. American Intergroup E1609. J Clin Oncol 2020;38:567–75.
178. Necchi A, Anichini A, Raggi D, Briganti A, Massa S, Lucianò R, 193. Ascierto PA, Del Vecchio M, Mandalá M, Gogas H, Arance AM, Dalle S,
et al. Pembrolizumab as neoadjuvant therapy before radical cystec- et al. Adjuvant nivolumab versus ipilimumab in resected stage IIIB-
tomy in patients with muscle-invasive urothelial bladder carcinoma C and stage IV melanoma (CheckMate 238): 4-year results from a
(PURE-01): an open-label, single-arm, phase II study. J Clin Oncol multicentre, double-blind, randomised, controlled, phase 3 trial.
2018;36:3353–60. Lancet Oncol 2020;21:1465–77.
179. Topalian SL, Bhatia S, Amin A, Kudchadkar RR, Sharfman WH, 194. Roudi R, Syn NL, Roudbary M. Antimicrobial peptides as biologic
Lebbé C, et al. Neoadjuvant nivolumab for patients with resectable and immunotherapeutic agents against cancer: a comprehensive
merkel cell carcinoma in the CheckMate 358 trial. J Clin Oncol overview. Front Immunol 2017;8:1320.
2020;38:2476–87. 195. Bommareddy PK, Shettigar M, Kaufman HL. Integrating oncolytic
180. Nanda R, Liu MC, Yau C, Shatsky R, Pusztai L, Wallace A, et  al. viruses in combination cancer immunotherapy. Nat Rev Immunol
Effect of pembrolizumab plus neoadjuvant chemotherapy on path- 2018;18:498–513.
ologic complete response in women with early-stage breast cancer: 196. Zamarin D, Holmgaard RB, Subudhi SK, Park JS, Mansour M,
an analysis of the ongoing phase 2 adaptively randomized I-SPY2 Palese P, et  al. Localized oncolytic virotherapy overcomes systemic
trial. JAMA Oncol 2020;6:676–84. tumor resistance to immune checkpoint blockade immunotherapy.
181. Mittendorf EA, Zhang H, Barrios CH, Saji S, Jung KH, Hegg R, et al. Sci Transl Med 2014;6:226ra32.
Neoadjuvant atezolizumab in combination with sequential nab- 197. Spranger S, Dai D, Horton B, Gajewski TF. Tumor-residing Batf3
paclitaxel and anthracycline-based chemotherapy versus placebo dendritic cells are required for effector T cell trafficking and adop-
and chemotherapy in patients with early-stage triple-negative breast tive T cell therapy. Cancer Cell 2017;31:711–23.
cancer (IMpassion031): a randomised, double-blind, phase 3 trial. 198. Theisen DJ, Ferris ST, Briseño CG, Kretzer N, Iwata A, Murphy KM,
Lancet 2020;396:1090–100. et  al. Batf3-dependent genes control tumor rejection induced by
182. Tolaney SM, Jerusalem G, Salgado R, Liu X, Chen T, Zhang H, et al. dendritic cells independently of cross-presentation. Cancer Immu-
A phase II trial of nivolumab (NIVO) + palbociclib (PAL) + anas- nol Res 2019;7:29–39.
trozole (ANA) in postmenopausal women and men with estrogen 199. Le Naour J, Zitvogel L, Galluzzi L, Vacchelli E, Kroemer G. Trial
receptor (ER)+/human epidermal growth factor 2 (HER2)- primary watch: STING agonists in cancer therapy. Oncoimmunology 2020;
breast cancer (BC): CheckMate 7A8. J Clin Oncol 2020;38:TPS1105. 9:1777624.
183. Loi S, McArthur HL, Harbeck N, Pusztai L, Delaloge S, Letrent K, 200. Le Naour J, Galluzzi L, Zitvogel L, Kroemer G, Vacchelli E. Trial
et al. A phase III trial of nivolumab with neoadjuvant chemotherapy watch: TLR3 agonists in cancer therapy. Oncoimmunology 2020;9:
and adjuvant endocrine therapy in ER+/HER2− primary breast can- 1771143.
cer: CheckMate 7FL. J Clin Oncol 2020;38:TPS604. 201. Frega G, Wu Q, Le Naour J, Vacchelli E, Galluzzi L, Kroemer G, et al.
184. Cardoso F, Bardia A, Andre F, Cescon DW, McArthur HL, Telli ML, Trial Watch: experimental TLR7/TLR8 agonists for oncological
et  al. KEYNOTE-756: randomized, double-blind, phase 3 study of indications. Oncoimmunology 2020;9:1796002.
pembrolizumab vs placebo combined with neoadjuvant chemo- 202. De Keersmaecker B, Claerhout S, Carrasco J, Bar I, Corthals J, Wil-
therapy and adjuvant endocrine therapy for high-risk, early-stage genhof S, et  al. TriMix and tumor antigen mRNA electroporated
estrogen receptor–positive, human epidermal growth factor recep- dendritic cell vaccination plus ipilimumab: link between T-cell acti-
tor 2–negative. J Clin Oncol 2019;37:TPS601. vation and clinical responses in advanced melanoma. J Immunother
185. Heymach J, Taube J, Mitsudomi T, Harpole D, Aperghis M, Trani L, Cancer 2020;8:e000329.
et al. P1.18-02 the AEGEAN phase 3 trial of neoadjuvant/adjuvant 203. Bommareddy PK, Patel A, Hossain S, Kaufman HL. Talimogene
durvalumab in patients with resectable stage II/III NSCLC. J Thorac laherparepvec (T-VEC) and other oncolytic viruses for the treatment
Oncol 2019;14:S625–6. of melanoma. Am J Clin Dermatol 2017;18:1–15.
186. Fernando HC, Yang J, Ferraro GL, Keller SM. Randomized, dou- 204. Andtbacka RHI, Collichio F, Harrington KJ, Middleton MR, Downey
ble-blind phase 3 study evaluating neoadjuvant platinum-based G, Öhrling K, et al. Final analyses of OPTiM: a randomized phase III
chemotherapy with perioperative pembrolizumab or placebo in trial of talimogene laherparepvec versus granulocyte-macrophage
resectable stage IIB or IIIA NSCLC: KEYNOTE-671. J Clin Oncol colony-stimulating factor in unresectable stage III-IV melanoma. J
2018;36:TPS8583. Immunother Cancer 2019;7:1–11.

856 | CANCER DISCOVERY 10TH ANNIVERSARY ISSUE April 2021 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
The Next Decade of Immune Checkpoint Therapy REVIEW

205. Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, et al. cd137 agonist, in patients with advanced cancer. Clin Cancer Res
The commensal microbiome is associated with anti-PD-1 efficacy in 2018;24:1816–23.
metastatic melanoma patients. Science 2018;359:104–8. 213. Rischin D, Groenland SL, Lim AML, Martin-Liberal J, Moreno V,
206. Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Trigo Perez JM, et al. Inducible T cell costimulatory (ICOS) receptor
Karpinets TV, et  al. Gut microbiome modulates response to anti- agonist, GSK3359609 (GSK609) alone and in combination with
PD-1 immunotherapy in melanoma patients. Science 2018;359: pembrolizumab (Pembro): preliminary results from INDUCE-1
97–103. expansion cohorts (EC) in head and neck squamous cell carcinoma
207. Suntharalingam G, Perry MR, Ward S, Brett SJ, Castello-Cortes A, (HNSCC). Ann Oncol 2019;30:v449–74.
Brunner MD, et al. Cytokine storm in a phase 1 trial of the anti-CD28 214. Mayes PA, Hance KW, Hoos A. The promise and challenges of immune
monoclonal antibody TGN1412. N Engl J Med 2006;355:1018–28. agonist antibody development in cancer. Nat Rev Drug Discov 2018;
208. Patel MR, Ellerton J, Infante JR, Agrawal M, Gordon M, Aljumaily R, 17:509–27.
et  al. Avelumab in metastatic urothelial carcinoma after platinum 215. Waite JC, Wang B, Haber L, Hermann A, Ullman E, Ye X, et  al.
failure (JAVELIN Solid Tumor): pooled results from two expansion Tumor-targeted CD28 bispecific antibodies enhance the antitumor
cohorts of an open-label, phase 1 trial. Lancet Oncol 2018;19:51–64. efficacy of PD-1 immunotherapy. Sci Transl Med 2020;12:eaba2325.
209. Chester C, Sanmamed MF, Wang J, Melero I. Immunotherapy 216. Zhang Q, Luo J, Wu S, Si H, Gao C, Xu W, et al. Prognostic and pre-
targeting 4-1BB: mechanistic rationale, clinical results, and future dictive impact of circulating tumor DNA in patients with advanced
strategies. Blood 2018;131:49–57. cancers treated with immune checkpoint blockade. Cancer Discov
210. Choi Y, Shi Y, Haymaker CL, Naing A, Ciliberto G, Hajjar J. T-cell 2020;10:1842–53.
agonists in cancer immunotherapy. J Immunother Cancer 2020;8: 217. Wells DK, van Buuren MM, Dang KK, Hubbard-Lucey VM, Shee-
e000966. han KCF, Campbell KM, et  al. Key parameters of tumor epitope
211. Segal NH, Logan TF, Hodi FS, McDermott D, Melero I, Hamid O, et al. immunogenicity revealed through a consortium approach improve
Results from an integrated safety analysis of urelumab, an agonist anti- neoantigen prediction. Cell 2020;183:818–34.
CD137 monoclonal antibody. Clin Cancer Res 2017;23:1929–36. 218. Alspach E, Lussier DM, Miceli AP, Kizhvatov I, DuPage M, Luoma AM,
212. Segal NH, He AR, Doi T, Levy R, Bhatia S, Pishvaian MJ, et  al. et al. MHC-II neoantigens shape tumour immunity and response to
Phase i study of single-agent utomilumab (PF-05082566), a 4-1bb/ immunotherapy. Nature 2019;574:696–701.

April 2021 CANCER DISCOVERY 10TH ANNIVERSARY ISSUE | 857

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.
The Next Decade of Immune Checkpoint Therapy
Padmanee Sharma, Bilal A. Siddiqui, Swetha Anandhan, et al.

Cancer Discov 2021;11:838-857.

Updated version Access the most recent version of this article at:
http://cancerdiscovery.aacrjournals.org/content/11/4/838

Cited articles This article cites 213 articles, 58 of which you can access for free at:
http://cancerdiscovery.aacrjournals.org/content/11/4/838.full#ref-list-1

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, use this link
http://cancerdiscovery.aacrjournals.org/content/11/4/838.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's
(CCC)
Rightslink site.

Downloaded from cancerdiscovery.aacrjournals.org on April 9, 2021. © 2021 American Association for Cancer
Research.

You might also like