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 resistance, optimizing patient selection, improving management of immune-related adverse events, and identifying rational therapeutic combinations. These challenges will need a focused approach encompassing 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 minimizing toxicities through better patient selection and rational combinations.

Metastatic cancers remain incurable in most patients as conventional therapies that target tumor cells usually cannot provide a durable response. The discovery of immune checkpoints that regulate immune responses transformed cancer care, offering long-term clinical responses and the possibility of cure in many more patients with metastatic cancer (1). In 2011, the FDA approved the first immune checkpoint targeting agent, ipilimumab—a mAb targeting CTLA4—which opened the field of immune checkpoint therapy (ICT). Subsequently, mAbs blocking other checkpoints such as PD-1 or PD-L1 have received FDA approvals to treat a number of tumor types alone and in combination with other agents.

Despite the unprecedented durable clinical responses observed in subsets of patients, most patients do not respond, and some patients develop resistance to therapy after initial response. Furthermore, ICTs can result in life-threatening toxicities, known as immune-related adverse events (irAE). Therefore, research studies are ongoing to understand pathways of resistance to ICT and mechanisms of irAEs. In this review, we outline the fundamentals of ICT and the current state of ICT in the clinic. We propose that development of effective ICT combinations will require a reverse translational approach, in which hypotheses are generated from in-depth immune monitoring studies of patients' samples and then tested in appropriate preclinical models, thereby yielding important insights to guide rational therapeutic strategies 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. Bolstered by the latest technologies, in the next decade we will better understand how to combine ICT with agents targeting multiple other cell subsets and pathways in the innate and adaptive immune system. The future of ICT offers ongoing promise to improve outcomes in patients with cancer.

T cells are the soldiers of the immune response. Activation of T cells requires a coordinated and stepwise process involving two primary signals (Fig. 1; ref. 2). T cells express the T-cell receptor (TCR), which signals via the CD3 complex upon the interaction of the TCR with MHC plus its cognate peptide antigen on antigen-presenting cells (APC). T cells also constitutively express a costimulatory receptor, CD28, which binds to the B7 family of costimulatory molecules that are predominantly expressed on professional APCs. To activate or turn T cells “on,” both the TCR signal via CD3 (signal 1) and the CD28 costimulatory signal via B7 interactions (signal 2) are required (Fig. 1A). Interaction of APCs with T cells that bear the specific TCR for a given antigen leads to activation of T cells and T cell–mediated responses. T cells then differentiate and clonally expand, with subsequent formation of memory T cells (3).

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.

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.

Close modal

CD4 and CD8 T cells constitute the majority of T cells in the immune system. CD4 T cells differentiate into a variety of subtypes: Th1, Th2, Th17, follicular Th (Tfh), T regulatory (Treg), and Th9 cells. These subsets have distinct transcription factors that regulate their function and cytokine profiles, which affect immune responses that range from effector cell responses for tumor rejection to regulatory cell responses to suppress effector cells. These transcription factors include T-bet for Th1 cells, BCL6 for Tfh, RORγ for Th17 cells, and FOXP3 for Tregs. Although CD4 T cells are known as helper cells, accumulating evidence suggests a cytotoxic function demonstrated by certain CD4 T cells (4). CD8 T cells are typically regarded as a uniform population with cytotoxic function; however, recent studies have highlighted the diversity of the CD8 T-cell population (5–9).

In general, Th1 CD4 cells and CD8 T cells are the effector cells responsible for driving antitumor immune responses. However, these immune responses are controlled by multiple mechanisms. Upon T-cell activation via the TCR and CD28, T cells proliferate and produce cytokines, but also upregulate inhibitory molecules that attenuate T-cell activation (Fig. 1B). Therefore, T cells have limited time to function before they are reined in to prevent damage to normal cells. Multiple inhibitory mechanisms exist, both cell-intrinsic and cell-extrinsic, to control T-cell responses. The signals that control antitumor immunity and immune suppression are comprised of a tightly regulated process that forms the yin and yang of the immune response.

CTLA4-Mediated Negative Costimulation

CTLA4 is a canonical inhibitory molecule that is expressed immediately following engagement of TCR, with peak expression around 48 to 72 hours following T-cell activation. It bears structural homology to CD28 and binds to B7-1 (CD80) and B7-2 (CD86) molecules on APCs with higher affinity than CD28, leading to competitive inhibition of costimulatory CD28 signaling, thereby dampening T-cell signaling (10, 11). In early studies, an anti-CTLA4 antibody prevented CTLA4 from engaging B7, leading to prolonged T-cell responses and eradication of cancer cells (1). Similarly, the in vivo administration of anti-CTLA4 antibodies enhanced the antitumor effect of tumor-infiltrating T cells, leading to regression of established tumors and long-lived immunity in tumor models (1).

Antibody blockade of CTLA4 was proposed as a way to enhance T-cell responses in the setting of cancer. By removing the brakes from T cells, it was hypothesized that T cells would have a longer time to function, which would enable tumor rejection. Anti-CTLA4 opened an entire new field termed “ICT.” The success of anti-CTLA4 led to the identification of other pathways that regulate T-cell responses, including both costimulatory and coinhibitory pathways (Fig. 1C; refs. 1, 12).

PD-1–Mediated Negative Costimulation

The primary biological function of PD-1 is to maintain peripheral tolerance. PD-1 engages with its ligands PD-L1 and PD-L2, widely expressed on nonlymphoid tissues, primarily dampening T-cell activation in the periphery. PD-1 interacts with an SHP2 tyrosine phosphatase domain, leading to dephosphorylation of signaling molecules downstream of the TCR and inhibiting TCR-mediated activation of IL2 production and T-cell proliferation (13). Therefore, whereas CTLA4 inhibits signal 2 (CD28-B7), PD-1/PD-L1 inhibits signal 1 (TCR).

PD-L1 expression has been described on a variety of tumor types, and is directly correlated with poor prognosis in several cancers (14, 15). Preclinical models demonstrated that PD-1 blockade results in tumor rejection through reinvigoration of exhausted CD8 T cells that express PD-1, LAG3, and TIM3 and expansion of CD8 T cells, a hallmark of response to anti–PD-1 therapy (16).

Distinct Mechanisms of Action of CTLA4 and PD-1

Although both CTLA4 and PD-1 are inhibitory molecules that attenuate T-cell activation, they have different mechanisms of action, and inhibition of CTLA4 and PD-1 leads to distinct immune responses. Anti-CTLA4 primarily functions in T-cell priming and expands clonal diversity. Anti-CTLA4 predominantly affects CD4 T cells, with multiple studies identifying increased frequency of a Th1 subset of CD4 T cells that express inducible costimulator (ICOS) as a result of anti-CTLA4 therapy (17–19). Increased frequency of ICOS+ CD4 T cells in peripheral blood acts as a pharmacodynamic biomarker of anti-CTLA4 therapy (20), whereas increased frequency of ICOS+ CD4 T cells in tumor tissues, possibly in the context of tertiary lymphoid structures, tends to correlate with improved clinical outcomes with ICT (21). Anti-CTLA4 can also promote T-cell trafficking into immunologically “cold” tumors. Investigation into the mechanisms of antitumor activity of CTLA4 blockade demonstrated the contribution of both the effector T-cell and Treg compartments in preclinical models (22, 23). Of note, although CTLA4 blockade was shown to cause selective depletion of intratumoral Tregs through antibody-dependent cell-mediated cytotoxicity (ADCC) in murine models and ex vivo experiments on human tumor samples (24–26), subsequent studies comparing pretreatment and posttreatment tumor samples from patients who received anti-CTLA4 therapy, either as tremelimumab (IgG2 antibody) or as ipilimumab (IgG1 antibody), did not observe a decrease in intratumoral Tregs after therapy (27). It is possible that the expression of specific Fc receptors on APCs or other cell types may be limited in some human tumors, thereby affecting Treg depletion by anti-CTLA4 therapy in patients with cancer. In future studies, it may be important to ascertain the expression of specific Fc receptors within the tumor microenvironment (TME) to determine whether ICT antibodies will mediate ADCC to affect Treg frequency. In general, anti-CTLA4 expands CD4 effector T cells (Teff) with subsequent increase in the Teff/Treg ratio in responding tumors (22).

Anti–PD-1/PD-L1 antibodies predominantly affect exhausted CD8 T cells. Anti–PD-1/PD-L1 also does not appear to expand clonal diversity nor promote T-cell trafficking into tumors. Preclinical studies have shown that concurrent targeting of CTLA4 and PD-1/PD-L1 can improve therapeutic efficacy when compared with each alone (28–30). mAbs targeting CTLA4, PD-1, and PD-L1 have formed the foundation of effective ICT in patients and are now in widespread clinical use across a number of tumor types (31–34).

There are now more than 50 FDA approvals for ICTs in human cancers, which are briefly discussed in this section and summarized in Table 1. The clinically approved ICTs have been comprehensively reviewed in other sources (35, 36).

Table 1.

FDA-approved ICTs

Agent/targetIndicationYear of 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 chemotherapy 2015 
  4. Patients with metastatic RCC who have received prior therapy 2015 
  5. cHL that has relapsed or progressed after autologous hematopoietic stem cell transplantation and posttransplantation brentuximab vedotin or after ≥3 lines of therapy 2016 
  6. Recurrent or metastatic HNSCC with disease progression on or after platinum-based therapy 2016 
  7. Locally advanced or metastatic UC with disease progression during or following platinum-based chemotherapy 2017 
  8. Adult and pediatric patients with MSI-H or dMMR metastatic colorectal cancer that has progressed following chemotherapy 2017 
  9. HCC previously treated with sorafenib 2017 
 10. Melanoma with lymph node involvement (adjuvant) or metastatic disease following complete resection 2017 
 11. Unresectable advanced, recurrent, or metastatic esophageal squamous cell carcinoma after prior fluoropyrimidine- and platinum-based chemotherapy 2020 
 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 treatments 2015 
  3. First-line treatment of unresectable or metastatic melanoma 2015 
  4. Recurrent or metastatic HNSCC with disease progression on or after platinum-based chemotherapy 2016 
  5. First-line treatment of metastatic NSCLC, with high PD-L1 expression and no EGFR or ALK genomic tumor alterations 2016 
  6. Adult and pediatric refractory cHL or disease relapse after three or more prior lines of 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) 2017 
  7. First-line treatment of metastatic nonsquamous NSCLC, irrespective of PD-L1 expression (in combination with carboplatin/pemetrexed) 2017 
  8. Locally advanced or metastatic UC with disease progression on or after platinum-based 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 2017 
  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 adenocarcinoma, with PD-L1–positive tumors and disease progression on or after two or more prior lines of therapy 2017 
 11. Recurrent or metastatic cervical cancer, with tumor PD-L1 CPS ≥ 1 and disease progression on or after prior chemotherapy 2018 
 12. Primary mediastinal B-cell lymphoma, refractory disease, or relapse after two or more lines of therapy 2018 
 13. Metastatic, nonsquamous NSCLC (in combination with pemetrexed and cisplatin/carboplatin) 2018 
 14. Metastatic, squamous NSCLC (in combination with carboplatin and paclitaxel or nab-paclitaxel) 2018 
 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 PD-L1 CPS ≥ 1 2019 
 21. HNSCC, first-line treatment for metastatic or unresectable recurrent disease (in combination with platinum and fluorouracil) 2019 
 22. SCLC, metastatic, with disease progression on or after platinum-based chemotherapy and at least 1 other prior line of therapy 2019 
 23. Esophageal cancer, recurrent locally advanced or metastatic squamous cell, with tumor PD-L1 CPS ≥ 10 and disease progression after one or more prior lines of systemic therapy 2019 
 24. Endometrial carcinoma, advanced, non–MSI-H/dMMR with disease progression after prior systemic therapy and not candidate for curative surgery or radiation (in combination with lenvatinib) 2019 
 25. UC, high-risk, BCG-unresponsive, non–muscle invasive with carcinoma in situ with or without papillary tumors, ineligible for or declined cystectomy 2020 
 26. Recurrent or metastatic cutaneous squamous cell carcinoma not curable by surgery or radiation 2020 
 27. Unresectable or metastatic TMB-high (≥10 mut/Mb) solid tumors that have progressed on prior treatment with no satisfactory alternative treatment options. 2020 
 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 PD-L1 CPS ≥ 10 (in combination with chemotherapy) 2020 
4. Ipilimumab + nivolumab/anti-CTLA4 + anti–PD-1  1. BRAFV600 wild-type unresectable or metastatic melanoma 2015 
  2. BRAFV600 wild-type and BRAFV600 mutation–positive unresectable or metastatic melanoma 2016 
  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 chemotherapy regardless of PD-L1 status 2020 
  7. Unresectable malignant pleural mesothelioma 2020 
5. Durvalumab/anti–PD-L1  1. Unresectable stage III NSCLC with nonprogressive disease following concurrent platinum-based chemotherapy and radiotherapy 2018 
  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 platinum-based chemotherapy, either before or after surgery 2016 
  2. Metastatic NSCLC with disease progression during or following platinum-based chemotherapy, and progression on an FDA-approved targeted therapy if the tumor has EGFR or ALK gene abnormalities 2016 
  3. Locally advanced or metastatic UC not eligible for cisplatin chemotherapy whose tumors express PD-L1 2017 
  4. First-line, metastatic, nonsquamous SCLC without EGFR or ALK genomic alterations (in combination with bevacizumab, paclitaxel, and carboplatin) 2018 
  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 ≥1% (in combination with nab-paclitaxel) 2019 
  7. Metastatic NSCLC without EGFR or ALK genomic alterations (in combination with carboplatin/nab-paclitaxel) 2019 
  8. Metastatic NSCLC, first line, PD-L1 high (tumor cells ≥50% or immune cells ≥10%) without EGFR or ALK genomic alterations 2020 
  9. Metastatic or unresectable HCC, first line (in combination with bevacizumab) 2020 
 10. BRAFV600 mutation–positive unresectable or metastatic melanoma in combination with cobimetinib and vemurafenib 2020 
7. Avelumab/anti–PD-L1  1. Adult and pediatric patients with metastatic Merkel cell carcinoma, including those who have not received prior chemotherapy 2017 
  2. Locally advanced or metastatic UC with disease progression during or following platinum-based chemotherapy 2017 
  3. Advanced RCC, first line (in combination with axitinib) 2019 
  4. Maintenance treatment for locally advanced for metastatic UC that has not progressed with first-line platinum-containing chemotherapy 2020 
8. Cemiplimab/anti–PD-1  1. Metastatic or locally advanced cutaneous squamous cell carcinoma 2018 
Agent/targetIndicationYear of 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 chemotherapy 2015 
  4. Patients with metastatic RCC who have received prior therapy 2015 
  5. cHL that has relapsed or progressed after autologous hematopoietic stem cell transplantation and posttransplantation brentuximab vedotin or after ≥3 lines of therapy 2016 
  6. Recurrent or metastatic HNSCC with disease progression on or after platinum-based therapy 2016 
  7. Locally advanced or metastatic UC with disease progression during or following platinum-based chemotherapy 2017 
  8. Adult and pediatric patients with MSI-H or dMMR metastatic colorectal cancer that has progressed following chemotherapy 2017 
  9. HCC previously treated with sorafenib 2017 
 10. Melanoma with lymph node involvement (adjuvant) or metastatic disease following complete resection 2017 
 11. Unresectable advanced, recurrent, or metastatic esophageal squamous cell carcinoma after prior fluoropyrimidine- and platinum-based chemotherapy 2020 
 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 treatments 2015 
  3. First-line treatment of unresectable or metastatic melanoma 2015 
  4. Recurrent or metastatic HNSCC with disease progression on or after platinum-based chemotherapy 2016 
  5. First-line treatment of metastatic NSCLC, with high PD-L1 expression and no EGFR or ALK genomic tumor alterations 2016 
  6. Adult and pediatric refractory cHL or disease relapse after three or more prior lines of 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) 2017 
  7. First-line treatment of metastatic nonsquamous NSCLC, irrespective of PD-L1 expression (in combination with carboplatin/pemetrexed) 2017 
  8. Locally advanced or metastatic UC with disease progression on or after platinum-based 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 2017 
  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 adenocarcinoma, with PD-L1–positive tumors and disease progression on or after two or more prior lines of therapy 2017 
 11. Recurrent or metastatic cervical cancer, with tumor PD-L1 CPS ≥ 1 and disease progression on or after prior chemotherapy 2018 
 12. Primary mediastinal B-cell lymphoma, refractory disease, or relapse after two or more lines of therapy 2018 
 13. Metastatic, nonsquamous NSCLC (in combination with pemetrexed and cisplatin/carboplatin) 2018 
 14. Metastatic, squamous NSCLC (in combination with carboplatin and paclitaxel or nab-paclitaxel) 2018 
 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 PD-L1 CPS ≥ 1 2019 
 21. HNSCC, first-line treatment for metastatic or unresectable recurrent disease (in combination with platinum and fluorouracil) 2019 
 22. SCLC, metastatic, with disease progression on or after platinum-based chemotherapy and at least 1 other prior line of therapy 2019 
 23. Esophageal cancer, recurrent locally advanced or metastatic squamous cell, with tumor PD-L1 CPS ≥ 10 and disease progression after one or more prior lines of systemic therapy 2019 
 24. Endometrial carcinoma, advanced, non–MSI-H/dMMR with disease progression after prior systemic therapy and not candidate for curative surgery or radiation (in combination with lenvatinib) 2019 
 25. UC, high-risk, BCG-unresponsive, non–muscle invasive with carcinoma in situ with or without papillary tumors, ineligible for or declined cystectomy 2020 
 26. Recurrent or metastatic cutaneous squamous cell carcinoma not curable by surgery or radiation 2020 
 27. Unresectable or metastatic TMB-high (≥10 mut/Mb) solid tumors that have progressed on prior treatment with no satisfactory alternative treatment options. 2020 
 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 PD-L1 CPS ≥ 10 (in combination with chemotherapy) 2020 
4. Ipilimumab + nivolumab/anti-CTLA4 + anti–PD-1  1. BRAFV600 wild-type unresectable or metastatic melanoma 2015 
  2. BRAFV600 wild-type and BRAFV600 mutation–positive unresectable or metastatic melanoma 2016 
  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 chemotherapy regardless of PD-L1 status 2020 
  7. Unresectable malignant pleural mesothelioma 2020 
5. Durvalumab/anti–PD-L1  1. Unresectable stage III NSCLC with nonprogressive disease following concurrent platinum-based chemotherapy and radiotherapy 2018 
  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 platinum-based chemotherapy, either before or after surgery 2016 
  2. Metastatic NSCLC with disease progression during or following platinum-based chemotherapy, and progression on an FDA-approved targeted therapy if the tumor has EGFR or ALK gene abnormalities 2016 
  3. Locally advanced or metastatic UC not eligible for cisplatin chemotherapy whose tumors express PD-L1 2017 
  4. First-line, metastatic, nonsquamous SCLC without EGFR or ALK genomic alterations (in combination with bevacizumab, paclitaxel, and carboplatin) 2018 
  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 ≥1% (in combination with nab-paclitaxel) 2019 
  7. Metastatic NSCLC without EGFR or ALK genomic alterations (in combination with carboplatin/nab-paclitaxel) 2019 
  8. Metastatic NSCLC, first line, PD-L1 high (tumor cells ≥50% or immune cells ≥10%) without EGFR or ALK genomic alterations 2020 
  9. Metastatic or unresectable HCC, first line (in combination with bevacizumab) 2020 
 10. BRAFV600 mutation–positive unresectable or metastatic melanoma in combination with cobimetinib and vemurafenib 2020 
7. Avelumab/anti–PD-L1  1. Adult and pediatric patients with metastatic Merkel cell carcinoma, including those who have not received prior chemotherapy 2017 
  2. Locally advanced or metastatic UC with disease progression during or following platinum-based chemotherapy 2017 
  3. Advanced RCC, first line (in combination with axitinib) 2019 
  4. Maintenance treatment for locally advanced for metastatic UC that has not progressed with first-line platinum-containing chemotherapy 2020 
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-CTLA4 Monotherapy

Ipilimumab (anti-CTLA4) was the first ICT to gain FDA approval in 2011 for the treatment of metastatic melanoma, on the basis of phase III trials demonstrating improvements in overall survival compared with vaccine or chemotherapy (37, 38). Initial studies were challenging because of the paradoxical phenomenon of pseudoprogression, in which patients initially experienced tumor growth before subsequent regression. The benefit of ICT would not have been adequately captured by traditional metrics of tumor measurement (39–41). Survival therefore proved to be the most important indicator of ICT benefit. Indeed, approximately 20% of patients who received ipilimumab achieved long-term survival greater than three years, with survival of 10 years noted for some patients (38, 42). Although ipilimumab remains the only anti-CTLA4 therapy to have approval in many countries, including FDA approval, other mAbs are being studied, including tremelimumab and quavonlimab (43). New forms of anti-CTLA4 therapies are also under development, including glycoengineered antibodies that seek to enhance ADCC and prodrug forms with reduced systemic activity to improve safety (44).

Anti–PD-1/PD-L1 Monotherapy

In 2014, the first PD-1 inhibitors were approved for the treatment of unresectable or metastatic melanoma (45, 46). The PD-1/PD-L1 inhibitors are now approved across multiple tumor types, including skin, genitourinary, lung, head and neck, breast, lymphoma, gynecologic, and gastrointestinal cancers. These approvals are summarized in Table 1. The PD-1 mAbs include nivolumab (fully human IgG4), pembrolizumab (humanized IgG4), and cemiplimab (fully human IgG4). The PD-L1 mAbs include durvalumab (fully human IgG1κ), atezolizumab (humanized IgG1), and avelumab (fully human IgG1). Anti–PD-1 therapies have also received approval for treatment of patients with cancer who are diagnosed with tumors that have microsatellite instability–high (MSI-H)/mismatch-repair deficiency (dMMR; refs. 47, 48) or high tumor mutational burden (TMB-high) regardless of tumor type (49). In addition, anti–PD-1/PD-L1 therapies were also approved by the FDA as maintenance therapy, which is administered after initial treatment of locally advanced or metastatic disease. Clinical benefit with maintenance ICT in specific settings was demonstrated in patients with non–small cell lung cancer (NSCLC; ref. 50), extensive-stage small cell lung cancer (51, 52), and bladder cancer (53), leading to FDA approvals in these tumor types.

Combination of Anti-CTLA4 and Anti–PD-1/PD-L1

Despite the promising responses with ICT monotherapies, these responses are observed in only 20% to 30% of treated patients. Therefore, combination therapy was pursued in the clinic based on preclinical data demonstrating that anti-CTLA4 and anti–PD-1/PD-L1 function through distinct pathways, and combined inhibition enhanced antitumor responses in murine models (28).

Multiple clinical trials with combined blockade of CTLA4 and PD-1/PD-L1 demonstrated higher response rates than either monotherapy, in different cancer types including melanoma (54, 55), hepatocellular carcinoma (56), renal cell carcinoma (RCC; refs. 57, 58), NSCLC (59), malignant pleural mesothelioma (60), and colorectal cancer (MSI-H/dMMR; ref. 61). Long-term follow-up has demonstrated durable survival benefit for a subset of patients with melanoma and RCC, and combination therapy with ICT is under active investigation for a number of tumor types (55, 58).

Combination of ICT with Other Therapeutic Agents

The quest to improve response rates has also prompted combinations with other established cancer drugs, for example, chemotherapy, which is hypothesized to kill immunosuppressive cells in the TME while increasing exposure to tumor antigens via the induction of tumor cell death, thereby enhancing recognition and elimination of tumor cells by the host immune system (62). Toward this end, combinations of ICTs with concurrent and/or sequential chemotherapy are under extensive investigation, with phase III clinical data demonstrating overall survival and/or progression-free survival benefit in NSCLC (63–66), small cell lung cancer (51, 52), triple-negative breast cancer (67), bladder cancer (53), and head and neck squamous cell carcinoma (68). However, the impact of different chemotherapy agents on the immune response within the TME has not been fully elucidated. Similarly, improved clinical responses have been demonstrated with the combination of ICT and targeted therapy. Combinations of VEGF-targeted agents with anti–PD-1 have demonstrated clinical benefits in endometrial carcinoma (pembrolizumab plus lenvatinib; refs. 69, 70), RCC (pembrolizumab plus axitinib; nivolumab plus cabozantinib; avelumab plus axitinib), and hepatocellular carcinoma (atezolizumab plus bevacizumab; refs. 71–75). Recently, the addition of atezolizumab to targeted therapy was also shown to improve progression-free survival in BRAF-mutated melanoma (76).

Although these combination therapies offer the possibility of improving cancer-specific outcomes, they remain limited to specific tumor types and disease settings. For the field of cancer immunotherapy to move forward successfully, with improved clinical benefit for many more patients, we will require rational combination therapies, which should be based on a reasonable understanding of the mechanism of action of each agent and its impact on immune responses.

Immune-Related Adverse Events

As combination strategies are pursued, there is an increased risk of development of irAEs. Mitigation strategies, therefore, will require a deeper mechanistic understanding of the toxicities. These irAEs are distinct from the predictable side effects associated with conventional cancer therapies. The irAEs can affect any organ, most commonly skin, gastrointestinal tract, and endocrine system (including the thyroid, adrenal, and pituitary glands). The severity of irAEs can range from mild to even fatal. Clinical guidelines for the management of irAEs have been reviewed elsewhere (77). Although guidelines recommend high-dose corticosteroids for severe irAEs, often with prolonged tapers, this approach can be associated with serious iatrogenic side effects, highlighting the need for mechanistically informed, steroid-sparing immunosuppressive approaches. Early recognition and treatment of irAEs is essential to prevent progression to severe events, and major efforts are under way to develop biomarkers for earlier predictions of irAEs, including blood-based biomarkers such as CD8 T-cell clonal expansion, serum autoantibodies (reflective of B-cell response), and technology-based approaches with patient symptom reporting (78–81).

Published data indicate that nonfatal irAEs may be associated with improved outcomes in patients with cancer (82–96). Therefore, the development of strategies that can enable patients to continue on ICT, including the use of selective immunosuppressive agents, will be important as the field of ICT moves forward. For example, the recent development of a murine model of myocarditis, one of the most feared complications of ICT, provided a rationale for the use of CTLA4-Ig (abatacept), which blocks T-cell costimulation, although an open question remains as to whether this will interfere with antitumor immunity (97, 98). Similarly, in a study of patients with melanoma who developed colitis after ICT, single-cell RNA sequencing (scRNA-seq) and TCR sequencing identified tissue-resident colitis-associated CD8 T cells, consistent with the clinical observation that colitis symptoms tend to occur early after initiation of ICT, and also provided support for the use of the α4β7 antibody vedolizumab to decrease inflammatory immune responses in ICT-associated colitis (99).

Overall, as multiple mechanisms related to irAEs become clear, including both T cell– and B cell–mediated pathways, a major question will be to address whether the mechanisms of irAEs can successfully be decoupled from those of antitumor efficacy. Future ICT clinical trials may include components of selective, prophylactic immunosuppression for high-risk, biomarker-selected patients to prevent severe irAEs, or possibly include administration of targeted immunosuppression to enable continuation of ICT in patients.

To obtain a greater understanding of mechanisms of response and resistance to ICT, we propose that reverse translational studies on individual agents be undertaken, whereby each agent can be studied in small cohorts of patients, with longitudinal samples collected and analyzed thoroughly to generate hypotheses regarding immunologic mechanisms that can be tested in appropriate preclinical models, and with translation of the preclinical data to new clinical trials. These data sets may provide relevant information that can help with the design of rational clinical trials as well as avoid the pitfalls of failed clinical trials. We have adopted this integrated strategy to understand the pathways regulating immune responses and to guide rational combinatorial strategies, as outlined below.

Addressing Primary and Adaptive Resistance Mechanisms

Prostate cancer is considered a “cold” tumor due to the fact that it has few mutations, as compared with “hot” tumors such as melanoma and NSCLC. As a result of a low number of mutations, and therefore few antigens for T-cell recognition, as noted by scarce tumor-infiltrating T cells, prostate cancer has been predominantly resistant to treatment with ICT (100–102). The mechanisms by which prostate tumors exclude T cells or fail to elicit robust infiltration by T cells represent primary resistance mechanisms to ICT. Without tumor-infiltrating T cells, the blockade of a T cell–inhibitory pathway such as PD-1/PD-L1 is unlikely to provide clinical benefit. For example, a recent phase III clinical trial of the androgen receptor antagonist enzalutamide plus anti–PD-L1 failed to meet its primary endpoint of improved overall survival (103). Previous studies on human prostate cancers demonstrated few tumor-infiltrating T cells, with minimal to no expression of PD-1 or PD-L1, and lack of significant clinical benefit with anti–PD-1 monotherapy in patients with metastatic prostate cancer (104, 105). Therefore, the phase III combination study would have benefited from a smaller clinical trial to truly determine whether enzalutamide was capable of increasing tumor-infiltrating T cells with subsequent increase in PD-1 and PD-L1 expression within the TME (106). Similarly, a phase III combination study of anti-CTLA4 plus radiotherapy in patients with metastatic prostate cancer also failed to meet its primary endpoint (100, 107). The combination was conducted without understanding the immunologic impact of anti-CTLA4 or radiotherapy on the prostate TME, including sites of metastatic disease, which are predominantly bone metastases in patients with metastatic prostate cancer.

To address the immunologic impact of anti-CTLA4 on the prostate TME, a small clinical trial evaluated pretreatment and posttreatment tumor samples, which demonstrated that anti-CTLA4 was capable of increasing tumor-infiltrating T cells (105). However, anti-CTLA4 also led to an increase in compensatory inhibitory mechanisms, also known as adaptive resistance mechanisms, including increased frequency of cells expressing PD-1/PD-L1 and VISTA within the TME (105). VISTA is an inhibitory checkpoint that is highly expressed on myeloid cells [including macrophages, dendritic cells (DC), monocytes, neutrophils], and moderately expressed on T cells (108). This yin and yang of the immune response, where therapy may drive antitumor immune responses but also increase immunosuppressive mechanisms, is an important concept to take into consideration as new immunotherapeutic strategies are developed.

The data from this small clinical trial generated hypotheses regarding combination therapies with anti-CTLA4 plus anti–PD-1 and/or anti-VISTA. These hypotheses were tested in preclinical studies, and the data led to a new study with anti-CTLA4 plus anti–PD-1 (109). The new combination clinical trial elicited antitumor responses in some patients, but these responses were predominantly in patients with soft-tissue metastases as opposed to bone metastases (109). Immune monitoring data of posttreatment tumor samples collected from patients indicated that soft-tissue metastases were infiltrated by Th1 cells, whereas bone metastases were infiltrated by Th17 cells (110). The increased Th1 responses provided an explanation for why soft-tissue metastases responded better than bone metastases to ICT.

But why did ICT drive Th1 responses in soft-tissue metastases and Th17 responses in bone metastases? A preclinical model was designed to answer this question. Data from these experiments demonstrated high levels of IL6 and TGFβ in bone metastases as opposed to soft-tissue metastases, which are important cytokines for skewing T-cell response toward Th17 instead of Th1 (110). These preclinical data are now being used to develop a new combination therapy clinical trial with ICT plus an agent to target TGFβ.

On the basis of these data, the concept of organ-specific immune microenvironments will need to be addressed as the field of ICT moves forward in the next decade. Treatment of patients with metastatic disease will need to take into consideration whether certain organs, such as bone and liver, have distinct immune microenvironments that will require different immunotherapeutic strategies (111).

Immunosuppressive Myeloid Cells

The immune microenvironment is unique to each tumor type and plays a critical role in response and resistance to ICT. Therefore, it is important to understand the composition and phenotypic states of intratumoral immune cells of different tumor types to develop a tumor-specific ICT strategy. Analysis of the immune microenvironment of five different tumor types, including those that respond relatively well to ICT and those that do not, revealed that ICT-sensitive and ICT-resistant tumor types have distinct immune microenvironments. Interaction of tumor cells with the immune microenvironment guides the constitution and phenotypes of immune subpopulations.

Immunologically cold tumors, such as prostate cancer and glioblastoma multiforme (GBM), have few tumor-infiltrating T cells but higher frequency of immune-suppressive myeloid cells. Comparison of tumor-infiltrating lymphocytes (TIL) from GBM and multiple other tumor types led to the identification of a GBM-specific CD73hi myeloid cell population, which persists even after treatment with anti–PD-1 (112). CD73 is an extracellular ectonucleotidase that plays a role in the adenosine immunosuppressive pathway, inhibiting T-cell activation and proliferation. Preclinical studies with CD73 knockout mice showed improved survival with ICT compared with control mice, demonstrating a direct role for CD73 (112). These series of studies helped in the identification of CD73 as a specific immunotherapeutic target to improve antitumor immune responses to ICT in GBM, generating a combination strategy for a future clinical trial in GBM.

Other myeloid-specific targets, including chemokine receptors (CXCR4, CCR2, and CSF1R), intracellular mediators (IDO1 and PI3Kϒ), and proangiogenic agents (VEGF receptors, semaphorin 4D and angiopoietin-2), are also being explored in an attempt to limit the function of immunosuppressive (M2) macrophages while promoting the function of M1 macrophages, which facilitate antitumor immune responses (113).

The differentiation of macrophages into M1 or M2 subsets is a complex biological process that is being investigated. Many different signals, including cytokine and epigenetic signals, appear to play a role in driving macrophage differentiation. Interestingly, the process of phagocytosis, which is required for clearing tumor cells, also seems to play a role in driving M2 differentiation in an attempt to suppress immune responses and avoid further cell death (114), indicating the complexity of how immune responses are regulated. Detailed understanding of the signals and biological processes involved in the differentiation of macrophages will be needed to effectively target these cells.

Epigenetics and Immune Response

As outlined previously, the functional and phenotypic states of immune subpopulations play an important role in determining response to ICT. Cellular plasticity is often driven by epigenetic mechanisms. Although the role of the epigenetic machinery in regulating cancer cell plasticity has been characterized (115), we lack mechanistic insight into the epigenetic regulation of immune subsets, especially in the context of ICT.

It was demonstrated that ipilimumab induces the expression of EZH2, a key epigenetic enzyme that plays an important role in cellular plasticity (115–118). CD28 signaling leads to increased EZH2 expression on T cells (119). Anti-CTLA4 increases CD28 signaling, thereby leading to increased EZH2 expression on T cells. On the basis of these observations, a series of preclinical studies were designed that identified a potential role for EZH2 in driving adaptive resistance to anti-CTLA4 therapy. The combination of EZH2 inhibition with anti-CTLA4 therapy led to improved survival in the preclinical studies (116). These studies formed the basis for a clinical trial of the rational combination of an EZH1/2 inhibitor (DS3201) with ipilimumab in patients with genitourinary malignancies with primary resistance to ICT (NCT04388852). This represents the tip of the iceberg in terms of epigenetic regulation of immune cells, providing the foundation for the combination of epigenetic modulators with ICT (120).

It is clear that we currently have the tools at our disposal to interrogate human immune responses in great depth and we will need to take advantage of these tools, and combine the human data with appropriate preclinical models, to identify rational targets for combination strategies that will improve clinical outcomes for patients. Patient selection will also play an important part in developing effective treatments, and we will need to consider multiple factors, including both tumor-related factors and immune-related factors, to identify appropriate populations.

One of the major challenges in the field of ICT is the lack of a robust predictive biomarker for optimal patient selection. Traditionally, biomarkers have focused primarily on tumor-intrinsic factors or single immune-specific markers. As our understanding of determinants of antitumor immunity has improved, we appreciate that single biomarkers in isolation are insufficient. Combinatorial biomarker strategies that capture attributes of the host and tumor–immune ecosystem will be essential (Fig. 2; refs. 121–124).

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.

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.

Close modal

TMB and Neoantigen Load as Predictive Biomarkers

High TMB is associated with response to ICT in numerous cancer types, most notably melanoma and NSCLC (49). High TMB may contribute to the formation of neoantigens that are recognized by the immune system (125, 126). On a tissue-agnostic basis, anti–PD-1 (pembrolizumab) received FDA approval for the treatment of advanced pediatric and adult solid tumors that have high TMB (≥10 mutations/megabase). This is a promising step on which to build, and future studies on TMB as a predictive biomarker will address threshold validation as well as provide a deeper understanding of the immunogenicity of different mutational patterns (127).

Although high TMB led to FDA approval, it should be noted that certain tumor types with moderate TMB carry relatively high response rates to ICT, such as clear cell RCC (128, 129). In addition, low TMB does not preclude generation of effective antigen responses, nor does high TMB guarantee response to ICT (130–132). For example, it was shown that anti-CTLA4 therapy can induce effective neoantigen-specific T-cell responses in patients with prostate cancer, even in the background of low TMB (131). Therefore, one approach to enhance antigen-specific T-cell responses may be the combination 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” tumors such as glioblastoma and “hot” tumors such as melanoma (133–135). As vaccine therapies evolve to encompass appropriate formulations, adjuvants, and delivery methods that can elicit effective antitumor immune responses, it may be possible to take advantage of tumor mutations to generate neoantigen vaccines in combination with ICT for the development of successful therapeutic strategies.

DNA Damage Repair Pathways: MSI-H/dMMR and Homologous Repair Defects

There are fundamental links between DNA damage and immunogenicity. Tumors with DNA damage repair (DDR) defects, exemplified by MSI-H/dMMR status, were predicted to have high rates of neoantigen formation due to their high mutational load (approximately 10-fold compared with that of MMR-proficient tumors) and therefore higher rates of response to ICT (136). An initial study in patients with colorectal cancer demonstrated significantly higher rates of response to pembrolizumab in dMMR patients compared with MMR-proficient patients (47), a finding that was later extended to multiple tumor types (48). These data led to the tumor-agnostic approval of pembrolizumab for MSI-H/dMMR solid tumors (48).

Additionally, homologous repair defects, such as pathogenic mutations in BRCA1/2, can lead to enhanced immune response by multiple pathways including the release of cytosolic DNA, promotion of type I IFN signaling, and upregulation of PD-L1 (137–139). In a study of patients with urothelial carcinoma, higher response rates to anti–PD-1/PD-L1 antibodies were observed in patients with known or likely deleterious DDR alterations (140). Emerging evidence indicates that specific DDR mutations have differential effects on the tumor–immune microenvironment, and therefore mutations will need to be selected carefully as candidate predictive biomarkers (141). In all, an integrated approach incorporating events in specific driver genes and mutational signatures will be critical to develop additional genetic biomarkers of response (142).

PD-L1 Expression as a Predictive Biomarker

IHC evaluation of PD-L1 expression on tumor and/or immune cells is frequently performed to select patients for treatment with anti–PD-1/PD-L1 antibodies. A correlation between PD-L1 expression on tumor cells and clinical response is seen in certain circumstances (e.g., in NSCLC); however, in other cases, substantial responses can be observed in some patients with PD-L1–negative tumors, which highlights the difficulty of using PD-L1 as a single biomarker (34, 143, 144). PD-L1 expression is dynamic due to multiple factors, including T-cell responses and IFNγ signaling. Therefore, variations in the timing of biopsy of the tumor as well as the site of biopsy may influence whether PD-L1 expression is detected or not. Although a range of assays have been approved, there remains significant variation in antibodies, tissue handling techniques, and thresholds, which limits the uniform benefit of PD-L1 expression as an isolated biomarker, and efforts are under way to integrate PD-L1 expression with other biomarkers (described below; ref. 127).

TILs and Exclusion of TILs as Biomarkers of Response and Resistance

TILs have a long history in cancer immunotherapy, with demonstration in the 1980s that adoptive transfer of TILs derived from patients' melanoma tumors can lead to cancer regression (145). It was later shown that TILs, including CD8 T cells, correlated with clinical outcomes in multiple tumor types, including ovarian cancer and bladder cancer (146, 147). Subsequent research on the correlates of response of anti-CTLA4 therapy demonstrated that the extent of ICT-induced tumor necrosis was directly related to the ratio of intratumoral CD8 T cells to FOXP3+ Tregs (148). TILs are therefore under active investigation as a biomarker to guide selection of patients for treatment with ICT, although significant variability exists in definitions and thresholds (149, 150). In a prospective study, the AMADEUS trial (NCT03651271, ongoing) was designed to evaluate the benefit of classifying tumors into immunologically “hot” and “cold” tumors based on CD8+ T-cell density (≥15% and <15%, respectively), with anti–PD-1 (nivolumab) monotherapy administered as treatment for “hot” tumors, whereas anti-CTLA4 (ipilimumab) plus anti–PD-1 (nivolumab) combination is administered as treatment for “cold” tumors in an attempt to take advantage of the ability of anti-CTLA4 to drive T cells into “cold” tumors. In addition, the Immunoscore is an approach that aims to standardize and quantify CD3 and CD8 T cells infiltrating and surrounding a tumor (149, 151). However, due to variability in immune cell infiltration in different tumor types, its applicability across cancers is challenging. Multiple studies indicate that the location of T cells in the tumor (whether at the invasive margins or in tumor parenchyma) and the phenotype of the T cells (whether cells are memory-like, exhausted, and/or activated) will need to be taken into consideration (152–154).

In the same vein, T-cell exclusion is a mechanism of resistance to ICT, exemplified by “cold” tumors such as prostate cancer and pancreatic cancer, which have few T cells in the TME. Certain oncogenic pathways may enable tumors to exploit this mechanism of immune evasion. For example, tumor cell–intrinsic WNT/β-catenin activation has been shown to lead to exclusion of T cells within the TME (123, 155, 156). In addition, PTEN loss is also associated with increased signaling through the PI3K–AKT pathway and correlated with exclusion of CD8 T cells and poor clinical responses to ICT (124). Thus, patient selection for treatment and strategies to overcome T-cell exclusion in the TME will need to be considered in future ICT trials.

IFNγ Gene Signature as a Predictive Biomarker

IFNγ is a key cytokine produced by activated T cells, as well as natural killer (NK) and NK T cells, and is critical for effective antitumor immunity. Preclinical work using IFNγ-blocking antibody and murine models lacking essential IFNγ signaling molecules demonstrated a key role for IFNγ in immune surveillance and tumor rejection (157, 158). In addition, knockdown of IFNγ-receptor 1 led to impaired antitumor responses with anti-CTLA4 in a murine model of melanoma (159). Consistent with the preclinical data, patients with melanoma harboring genomic defects in IFNγ pathway genes did not respond to anti-CTLA4 (160). Also, patients with melanoma tumors harboring loss-of-function mutations in JAK1 and JAK2, downstream IFNγ signaling molecules, were resistant to anti–PD-1 therapy, highlighting the utility of IFNγ as a potential biomarker.

IFNγ induces a host of target genes that have been used to develop gene-expression signatures, which correlate with responses to ICT. The gene-expression signatures can be retrospectively labeled as “high” or “low” based on median cutoff values. Multiple studies have reported that tumors identified as having “high” expression of IFNγ-related gene signatures have better clinical responses to ICT (131, 161, 162). However, the gene panels used to design the IFN-gene signatures were not consistent across these studies. In addition, consistent with the yin and yang concept of immune responses, IFNγ drives expression of proteins such as PD-L1 and IDO1, which may act to suppress immune responses in the TME. Additional studies are ongoing to develop reproducible assays for detection of IFNγ-related genes and signatures that may be used as a prospective biomarker for selection of patients for ICT.

Tertiary Lymphoid Structures as a Predictive Biomarker

Tertiary lymphoid structures (TLS) have recently emerged as biomarkers of response to ICT across several tumor types, including lung cancer, melanoma, RCC, sarcoma, and urothelial cancer (163–167). TLS or TLS-like structures comprised of T cells, B cells, DCs, and other APCs may play a significant role in antitumor T-cell responses, likely due to further priming and activation of T cells within the TME, which may represent another important event in the cancer immunity cycle. In contrast to other biomarkers of response which give 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 types and forming the TLS, including CXCL13 (B cells) and IL2 (T cells; ref. 168).

TLS represent sites of interactions between multiple immune cell types and capture an ongoing antitumor response (168). Future studies using newer technologies will be essential to understand which cells are interacting within the TLS, and how they differ from the typical structures within lymph nodes. Finally, because TLS appear to be associated with responses to ICT across multiple tumor types, it will be important to define the specific mechanisms driving TLS formation to determine whether these mechanisms can be exploited to induce the formation of TLS in otherwise nonresponding tumors.

Combinatorial Biomarkers

Although many of the biomarkers described above, including tumor-intrinsic and host-specific variables, correlate with response to ICTs, many of the studies have been retrospective and need further validation through prospective clinical trials. Furthermore, there is a critical need to bridge preclinical mechanistic studies with clinical investigations and vice versa to develop reproducible predictive biomarkers. There is growing recognition that single biomarker strategies do not incorporate the dynamic and complex interaction of the tumor and host immune system and that combinatorial biomarkers will be essential to optimize patient selection. With inherent inter- and intratumor heterogeneity, a complex interplay of diverse cell types, and the continuously evolving tumor immune microenvironment, it is clear that the utility of isolated biomarkers is limited. A combination approach integrating multiple data sets across different platforms from diverse studies will be essential for developing more sensitive and robust biomarkers of response.

Recent studies demonstrated that the combination of existing biomarkers has better predictive capacity than single biomarkers. For example, TMB plus a T cell–inflamed gene-expression profile or TMB plus PD-L1 expression showed an improved prediction performance compared with single biomarkers (130). A recent study of more than 1,000 ICT-treated patients across seven tumor types showed that a multivariable predictor of response outperformed TMB alone (169). Such combinatorial biomarkers, tested in prospective clinical trials, will be key for optimal patient selection.

It was also recently demonstrated that two novel biomarkers correlate with response to anti–PD-1/PD-L1 therapies. A retrospective analysis identified mutations in ARID1A in tumor tissues and expression of the chemokine CXCL13 as predictive biomarkers for ICT (170). Preclinical studies demonstrated association of ARID1A and CXCL13 with antitumor immunity in mice (170). Importantly, the combination of ARID1A mutation plus CXCL13 showed improved predictive capacity compared with either of the single biomarkers alone (170). This retrospective study highlighted the importance of integrating tumor mutational status with the immune microenvironment to predict responses to ICT, which led to a prospective clinical trial to test ARID1A mutation plus CXCL13 as a combinatorial biomarker strategy for patient selection.

The field of ICT has expanded across many different tumor types, with improved benefit for patients with cancer. In addition, as the field continues to highlight other pathways that can be targeted, there are now a vast number of combinatorial approaches than can be tested in patients. Therefore, it will be important to efficiently design future studies with attention to the mechanisms of action of each agent as well as an understanding of resistance mechanisms that evolve over time to control immune responses. The field of ICT is still growing, but as we gain knowledge in multiple areas, as outlined below, the field will mature to provide clinical benefit for even more patients with cancer (Fig. 3).

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.

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.

Close modal

Integration of ICT with Other Therapies

Over the past decade, ICT has become established as the fourth pillar of cancer therapy, along with chemotherapy/targeted therapy, radiotherapy, and surgery. We have seen some success in combining ICT with chemotherapy and targeted therapy. The next decade will provide scientific insights into integrating ICT with these pillars of cancer care to benefit more patients. Exciting preclinical data on radiotherapy plus ICT should provide insight into translating it to clinical practice. Although surgical options in the metastatic setting are extremely limited, we are exploring combinations of ICT with surgery in the metastatic setting. These studies will broaden the field of ICT across all aspects of oncology as a unifying foundation for rational combination strategies with chemotherapy, targeted therapy, radiotherapy, and surgery. In addition, a number of other potential candidates such as other immune checkpoints and costimulatory receptors, epigenetic modulators, and metabolic targets hold promise for improving the durable clinical responses seen with ICT.

Moving from Metastatic to Earlier Disease Stages

Treatment of patients with earlier stages of disease, with therapy given in either the neoadjuvant or adjuvant setting, has the potential to prevent disease relapse and lead to improved survival. Neoadjuvant ICT offers the possibility of surgical downstaging and provides ample tissue for examination of an intact TME, potentially yielding insight into posttreatment changes that can inform subsequent clinical trials. The first neoadjuvant ICT trial was conducted in 2006 and provided safety data indicating that ICT can be given prior to surgery (17). The study was conducted in patients with localized bladder cancer who received anti-CTLA4 before proceeding to cystectomy (17). This study was also the first ICT trial in patients with bladder cancer, which provided critical data demonstrating antitumor responses in patients with bladder cancer, thereby leading to large clinical trials with ICT in patients with metastatic bladder cancer, with subsequent FDA approval. In addition, the immune monitoring studies performed on the trial highlighted the reverse translational approach and identified ICOS+ CD4 T cells as a novel subset of T cells associated with anti-CTLA4 therapy (17, 18, 171, 172). The trial also led to the first combination ICT neoadjuvant trial with anti-CTLA4 plus anti–PD-L1 in patients with bladder cancer, which demonstrated clinical responses and correlation of TLS with responses (21). Clinical trials with ICT in the neoadjuvant setting have now demonstrated clinical benefit in multiple tumor types, including melanoma (173, 174), Merkel cell carcinoma (175), NSCLC (176), MMR-deficient colorectal cancer (177), urothelial carcinoma (21, 178), and breast cancer (179–181). Additional studies are being planned, especially as investigators consider whether to implement survival or other clinical endpoints, including pathologic complete responses, which will enable faster readout and review for possible approval (182–188).

Administration of ICT in the neoadjuvant setting enables T and B cells to have access to the antigens that are present on tumor tissues, which is in contrast to adjuvant studies where ICT is given after tumor tissues have been removed, thereby limiting the amount of tumor antigens that T and B cells encounter. Nonetheless, adjuvant ICT has been successful in high-risk melanoma, which led to FDA approval (189–193). A number of additional studies are currently ongoing and will provide additional insight into the use of ICT in the adjuvant setting.

Targeting Innate Immune Responses

Although much attention has traditionally focused on adaptive immunity, there is growing interest in the role of innate immune responses to boost overall antitumor immunity. The innate immune system is composed of NK cells, dendritic cells, macrophages, monocytes, neutrophils, eosinophils, basophils, and mast cells. Therapeutic approaches include stimulating innate responses with microbial peptides or oncolytic viruses, which have previously been shown to enhance local and systemic antitumor immunity in combination with ICT and may be able to improve payload to enhance agonist signals (194–196). The presence of tumor-infiltrating DCs expressing the transcription factor BATF as well as the CD103 marker has been shown to be essential for presentation of neoantigens and activation of cytotoxic T cells in the TME (197, 198). Therefore, approaches that enhance and/or activate these DCs may improve local antitumor responses. Systemic approaches to stimulating innate immunity, such as via agonism of stimulator of interferon response cGAMP interactor 1 (STING) or Toll-like receptors (TLR), have been challenging (199–201). Therefore, intratumoral delivery rather than systemic delivery of agents to target innate immune cells may provide an avenue to ensure presentation of tumor-specific antigens, enable effective T-cell responses, and also reduce systemic toxicities. Intratumoral agents under investigation in clinical trials include mRNAs encoding CD70, CD40 ligand, and TLR4 (202) and oncolytic viruses such as talimogene laherparepvec, or T-VEC (203, 204), which is FDA- approved for melanoma and is being explored in a number of clinical trials. However, successful DC activation in a suppressive TME continues to remain a challenge and needs to be further investigated.

Targeting Microbiome to Drive Innate and Adaptive Immune Responses

Furthermore, as our understanding of host effects on the immune system has grown, it is clear that the gut microbiome exerts influence over the effectiveness of ICTs (127). Responses to ICTs have been correlated with commensal gut flora, and there appear to be substantial differences in the diversity and composition in patients who respond to ICT versus nonresponders (205, 206). Challenges in investigating the gut microbiome include differences in sampling, analysis platforms, and population heterogeneity, though these can be addressed with the development of standard sequencing and analytic tools. Manipulation of the gut microbiome, such as through fecal microbiota transplantation, antibiotic therapy, or diet, may serve as an adjunct strategy to enhance responses to ICT. These strategies are still in early stages of being explored but will provide important data that will affect treatment with ICT.

Driving Agonist Signals for Combination Therapy with ICT

On the basis of our understanding of T-cell activation, there is growing interest in driving agonist signals via costimulatory receptors. However, clinical experience with a CD28 superagonist that resulted in severe toxicities has led the field to proceed with caution as additional agonist antibodies are evaluated (207). Both OX40 and 4-1BB/CD137 agonists have been tested in clinical trials. Preclinical work with OX40 and 4-1BB showed efficacy; however, a phase Ib study of an OX40 agonist combined with atezolizumab demonstrated safety but limited efficacy in patients (208–210). In addition, phase I studies with CD137-directed mAb urelumab caused significant elevation of liver enzymes (211), and a second anti-CD137 antibody, utomilumab, was well tolerated but had limited efficacy (212). These studies highlight significant knowledge gaps regarding the use of agonist antibodies in terms of understanding expression of the target proteins, sequencing of the agonist antibody with ICT and choice of ICT agent (anti-CTLA4 vs. anti–PD-1/PD-L1). Currently, a clinical trial with anti-CTLA4 plus an agonist anti-ICOS antibody is ongoing (NCT03989362) based on preclinical data demonstrating enhanced antitumor immune responses in murine models of melanoma and prostate cancer (172). In addition, the ICOS agonist GSK3359609 is also under study in a phase III clinical trial (NCT04128696; ref. 213).

The development of immune agonists faces a number of challenges, including target affinity, epitope selection, receptor occupancy, Fc gamma receptor interactions, antibody isotype, and appropriate in vitro assays to clearly distinguish between an antagonist antibody with activity against a costimulatory molecule expressed on immunosuppressive cells versus an agonist antibody with activity against the same costimulatory molecule expressed on effector cells (214). As a result, the development of an agonist antibody to target T-cell responses may require additional experimental steps and careful immune monitoring studies of patients to help design appropriate combination trials with current ICT agents.

As the field builds on current data with agonist antibodies, novel approaches are being developed to deliver agonist signals in the TME. For example, bispecific antibodies are being explored, including a bispecific antibody to target CD28 plus a tumor-associated antigen (NCT03972657; ref. 215). This approach of targeting the costimulatory pathway within the TME, as opposed to systemic targeting, may provide a safer way to provide agonist signals to T cells. As these processes are optimized, costimulatory receptor targeting is becoming a feasible strategy to enhance the efficacy of ICT.

Incorporation of Newer Technologies

In the future, spatiotranscriptomic tools such as Digital Spatial Profiling and CO-Detection by indEXing (CODEX) may allow us to decipher the cell types and their interactions within the tumor. In addition, the incorporation of single-cell high-throughput technologies into clinical and preclinical studies will enable better characterization of cell subsets regulating antitumor responses. Assessing single-cell transcriptomics, proteomics, and chromatin state through scRNA-seq, Cellular Indexing of Transcriptomes and Epitopes by Sequencing (CITE-seq), and single-cell Assay for Transposase-Accessible Chromatin sequencing (scATAC-seq) will enable identification of cellular identity at a single-cell level. Furthermore, integration of various platforms assisted by novel bioinformatic tools will allow us to understand the regulatory pathways governing response and resistance to ICT. In addition, integration of longitudinal blood-based assays such as circulating-tumor DNA (ctDNA; ref. 216), which are easier to implement and less invasive as opposed to longitudinal tumor biopsies, is under investigation as a method to monitor responses to ICT.

Additionally, the development of better bioinformatics algorithms that help with neoantigen prediction will enable identification of more relevant therapeutic targets. Progress in the prediction of shared MHC class I antigens in clinical samples has been exciting. A recent study developed a pipeline to determine tumor epitope immunogenicity that might enable prediction of effective antitumor responses with much higher accuracy than conventional methods (217). However, although developing prediction algorithms for class I antigens is necessary, it has recently been shown that MHC class II neoantigens are also involved in effective tumor responses and will need to be considered (218).

The successful implementation of these technologies will require close collaboration between computational biologists and immunologists to avoid simplification of the complex biological and functional processes of immune responses, thereby maintaining fidelity to the dynamic nature of cell subsets within the immune system. For example, PD-1 expression on a T cell may represent an “activated” T cell or an “exhausted” T cell. The simple identification of PD-1 through a bioinformatics approach may not provide sufficient understanding regarding the complexity of the T-cell response. Therefore, appropriate functional studies or other immunologic assays may be necessary to identify additional biomarkers that should be included in the bioinformatic analyses for a more accurate definition of a given immune cell state. Careful attention to these types of details will be necessary for advancing the field of ICT.

Over the next decade, the field of cancer immunotherapy will need to grow vertically, to gain a deeper understanding of different immune cell subsets and pathways that regulate their responses, as well as horizontally, to integrate with other fields, including cancer biology, epigenetics, computational biology, and many others. ICT will also need to expand across all of oncology for the development of combination strategies, including combinations with surgery, radiotherapy, chemotherapy, targeted therapy, and other immunotherapy agents such as novel immune checkpoints and chimeric antigen receptor T cells. As the field continues to grow, it will need to remain grounded in the strength of basic immunology while it fosters close collaborations with other scientific disciplines. The future of ICT is promising, with ample opportunity to further improve outcomes for patients with cancer.

P. Sharma reports personal fees from Jounce Therapeutics, Oncolytics, Earli, Glympse Bio, Lava Therapeutics, Lytix, Phenomics, Codiak, Constellation, Achelois, Dragonfly, BioAtla, Infinity Pharma, Hummingbird, and Polaris outside the submitted work; in addition, P. Sharma has a patent for ARID1A plus CXCL13 as combinatorial biomarker pending and a patent for combination of anti-CTLA4 plus agonist targeting of ICOS for improved antitumor immune responses licensed to Jounce Therapeutics. S.K. Subudhi reports personal fees from Dendreon, Kahr Medical, Ltd., Cancer Expert Now, Inc., Bayer Healthcare Pharmaceuticals, Apricity Heatlh, LLC, Dava Oncology, Bristol-Meyers Squibb, Amgen, Inc., and Exelixis, Inc. outside the submitted work. J.P. Allison reports personal fees from Jounce Therapeutics, Codiak Biosciences, Achelois, Lava Therapeutics, Lytix, Earli, Phenomics, Dragonfly, Hummingbird, ImaginAb, Forty-Seven, and Polaris outside the submitted work. No disclosures were reported by the other authors.

1.
Leach
DR
,
Krummel
MF
,
Allison
JP
. 
Enhancement of antitumor immunity by CTLA-4 blockade
.
Science
1996
;
271
:
1734
6
.
2.
Sharma
P
,
Allison
JP
. 
The future of immune checkpoint therapy
.
Science
2015
;
348
:
56
61
.
3.
Smith-Garvin
JE
,
Koretzky
GA
,
Jordan
MS
. 
T cell activation
.
Annu Rev Immunol
2009
;
27
:
591
619
.
4.
Śledzińska
A
,
Vila de Mucha
M
,
Bergerhoff
K
,
Hotblack
A
,
Demane
DF
,
Ghorani
E
, et al
Regulatory T cells restrain interleukin-2- and Blimp-1-dependent acquisition of cytotoxic function by CD4(+) T cells
.
Immunity
2020
;
52
:
151
66
.
5.
Khan
O
,
Giles
JR
,
McDonald
S
,
Manne
S
,
Ngiow
SF
,
Patel
KP
, et al
TOX transcriptionally and epigenetically programs CD8(+) T cell exhaustion
.
Nature
2019
;
571
:
211
8
.
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 differentiation
.
Nature
2019
;
571
:
270
4
.
7.
Chen
Z
,
Ji
Z
,
Ngiow
SF
,
Manne
S
,
Cai
Z
,
Huang
AC
, et al
TCF-1-centered transcriptional network drives an effector versus exhausted CD8 T cell-fate decision
.
Immunity
2019
;
51
:
840
55
.
8.
Hudson
WH
,
Gensheimer
J
,
Hashimoto
M
,
Wieland
A
,
Valanparambil
RM
,
Li
P
, et al
Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1(+) stem-like CD8(+) T cells during chronic infection
.
Immunity
2019
;
51
:
1043
58
.
9.
McLane
LM
,
Abdel-Hakeem
MS
,
Wherry
EJ
. 
CD8 T cell exhaustion during chronic viral infection and cancer
.
Annu Rev Immunol
2019
;
37
:
457
95
.
10.
Alegre
ML
,
Noel
PJ
,
Eisfelder
BJ
,
Chuang
E
,
Clark
MR
,
Reiner
SL
, et al
Regulation of surface and intracellular expression of CTLA4 on mouse T cells
.
J Immunol
1996
;
157
:
4762
70
.
11.
Takahashi
T
,
Tagami
T
,
Yamazaki
S
,
Uede
T
,
Shimizu
J
,
Sakaguchi
N
, et al
Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4
.
J Exp Med
2000
;
192
:
303
10
.
12.
Krummel
MF
,
Allison
JP
. 
CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation
.
J Exp Med
1995
;
182
:
459
65
.
13.
Okazaki
T
,
Honjo
T
. 
PD-1 and PD-1 ligands: from discovery to clinical application
.
Int Immunol
2007
;
19
:
813
24
.
14.
Inman
BA
,
Sebo
TJ
,
Frigola
X
,
Dong
H
,
Bergstralh
EJ
,
Frank
I
, et al
PD-L1 (B7-H1) expression by urothelial carcinoma of the bladder and BCG-induced granulomata: associations with localized stage progression
.
Cancer
2007
;
109
:
1499
505
.
15.
Hamanishi
J
,
Mandai
M
,
Iwasaki
M
,
Okazaki
T
,
Tanaka
Y
,
Yamaguchi
K
, et al
Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer
.
Proc Natl Acad Sci U S A
2007
;
104
:
3360
5
.
16.
Im
SJ
,
Hashimoto
M
,
Gerner
MY
,
Lee
J
,
Kissick
HT
,
Burger
MC
, et al
Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy
.
Nature
2016
;
537
:
417
21
.
17.
Carthon
BC
,
Wolchok
JD
,
Yuan
J
,
Kamat
A
,
Ng Tang
DS
,
Sun
J
, et al
Preoperative CTLA-4 blockade: tolerability and immune monitoring in the setting of a presurgical clinical trial
.
Clin Cancer Res
2010
;
16
:
2861
71
.
18.
Liakou
CI
,
Kamat
A
,
Tang
DN
,
Chen
H
,
Sun
J
,
Troncoso
P
, et al
CTLA-4 blockade increases IFNgamma-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients
.
Proc Natl Acad Sci U S A
2008
;
105
:
14987
92
.
19.
Chen
H
,
Liakou
CI
,
Kamat
A
,
Pettaway
C
,
Ward
JF
,
Tang
DN
, et al
Anti-CTLA-4 therapy results in higher CD4 +ICOS hi T cell frequency and IFNγ levels in both nonmalignant and malignant prostate tissues
.
Proc Natl Acad Sci U S A
2009
;
106
:
2729
34
.
20.
Ng Tang
D
,
Shen
Y
,
Sun
J
,
Wen
S
,
Wolchok
JD
,
Yuan
J
, et al
Increased frequency of ICOS+ CD4 T cells as a pharmacodynamic biomarker for anti-CTLA-4 therapy
.
Cancer Immunol Res
2013
;
1
:
229
34
.
21.
Gao
J
,
Navai
N
,
Alhalabi
O
,
Siefker-Radtke
A
,
Campbell
MT
,
Tidwell
RS
, et al
Neoadjuvant PD-L1 plus CTLA-4 blockade in patients with cisplatin-ineligible operable high-risk urothelial carcinoma
.
Nat Med
2020
;
26
:
1845
51
.
22.
Quezada
SA
,
Peggs
KS
,
Curran
MA
,
Allison
JP
. 
CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells
.
J Clin Invest
2006
;
116
:
1935
45
.
23.
Peggs
KS
,
Quezada
SA
,
Chambers
CA
,
Korman
AJ
,
Allison
JP
. 
Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies
.
J Exp Med
2009
;
206
:
1717
25
.
24.
Wing
K
,
Onishi
Y
,
Prieto-Martin
P
,
Yamaguchi
T
,
Miyara
M
,
Fehervari
Z
, et al
CTLA-4 control over Foxp3+ regulatory T cell function
.
Science
2008
;
322
:
271
5
.
25.
Read
S
,
Greenwald
R
,
Izcue
A
,
Robinson
N
,
Mandelbrot
D
,
Francisco
L
, et al
Blockade of CTLA-4 on CD4+CD25+ regulatory T cells abrogates their function in vivo
.
J Immunol
2006
;
177
:
4376
83
.
26.
Romano
E
,
Kusio-Kobialka
M
,
Foukas
PG
,
Baumgaertner
P
,
Meyer
C
,
Ballabeni
P
, et al
Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients
.
Proc Natl Acad Sci U S A
2015
;
112
:
6140
5
.
27.
Sharma
A
,
Subudhi
SK
,
Blando
J
,
Scutti
J
,
Vence
L
,
Wargo
J
, et al
Anti-CTLA-4 immunotherapy does not deplete Foxp3 þ regulatory T cells (Tregs) in human cancers
.
Clin Cancer Res
2019
;
25
:
1233
8
.
28.
Curran
MA
,
Montalvo
W
,
Yagita
H
,
Allison
JP
. 
PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors
.
Proc Natl Acad Sci U S A
2010
;
107
:
4275
80
.
29.
Wei
SC
,
Levine
JH
,
Cogdill
AP
,
Zhao
Y
,
Anang
NAAS
,
Andrews
MC
, et al
Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade
.
Cell
2017
;
170
:
1120
33
.
30.
Wei
SC
,
Sharma
R
,
Anang
N-AAS
,
Levine
JH
,
Zhao
Y
,
Mancuso
JJ
, et al
Negative co-stimulation constrains T cell differentiation by imposing boundaries on possible cell states
.
Immunity
2019
;
50
:
1084
98
.
31.
Berger
R
,
Rotem-Yehudar
R
,
Slama
G
,
Landes
S
,
Kneller
A
,
Leiba
M
, et al
Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies
.
Clin Cancer Res
2008
;
14
:
3044
51
.
32.
Brahmer
JR
,
Drake
CG
,
Wollner
I
,
Powderly
JD
,
Picus
J
,
Sharfman
WH
, et al
Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates
.
J Clin Oncol
2010
;
28
:
3167
75
.
33.
Brahmer
JR
,
Tykodi
SS
,
Chow
LQM
,
Hwu
W-J
,
Topalian
SL
,
Hwu
P
, et al
Safety and activity of anti-PD-L1 antibody in patients with advanced cancer
.
N Engl J Med
2012
;
366
:
2455
65
.
34.
Topalian
SL
,
Hodi
FS
,
Brahmer
JR
,
Gettinger
SN
,
Smith
DC
,
McDermott
DF
, et al
Safety, activity, and immune correlates of anti-PD-1 antibody in cancer
.
N Engl J Med
2012
;
366
:
2443
54
.
35.
Hargadon
KM
,
Johnson
CE
,
Williams
CJ
. 
Immune checkpoint blockade therapy for cancer: an overview of FDA-approved immune checkpoint inhibitors
.
Int Immunopharmacol
2018
;
62
:
29
39
.
36.
Vaddepally
RK
,
Kharel
P
,
Pandey
R
,
Garje
R
,
Chandra
AB
. 
Review of indications of FDA-approved immune checkpoint inhibitors per NCCN guidelines with the level of evidence
.
Cancers
2020
;
12
:
738
.
37.
Hodi
FS
,
O'Day
SJ
,
McDermott
DF
,
Weber
RW
,
Sosman
JA
,
Haanen
JB
, et al
Improved survival with ipilimumab in patients with metastatic melanoma
.
N Engl J Med
2010
;
363
:
711
23
.
38.
Robert
C
,
Thomas
L
,
Bondarenko
I
,
O'Day
S
,
Weber
J
,
Garbe
C
, et al
Ipilimumab plus dacarbazine for previously untreated metastatic melanoma
.
N Engl J Med
2011
;
364
:
2517
26
.
39.
Seymour
L
,
Bogaerts
J
,
Perrone
A
,
Ford
R
,
Schwartz
LH
,
Mandrekar
S
, et al
iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics
.
Lancet Oncol
2017
;
18
:
e143
52
.
40.
Pignon
JC
,
Jegede
O
,
Shukla
SA
,
Braun
DA
,
Horak
CE
,
Wind-Rotolo
M
, et al
irRECIST for the evaluation of candidate biomarkers of response to nivolumab in metastatic clear cell renal cell carcinoma: analysis of a phase II prospective clinical trial
.
Clin Cancer Res
2019
;
25
:
2174
84
.
41.
Ayoub
M
,
Eleneen
Y
,
Colen
RR
. 
Cancer imaging in immunotherapy
.
In
:
Naing
A
,
Hajjar
J
,
editors
.
Immunotherapy
.
Cham
:
Springer International Publishing
; 
2020
.
p.
309
24
.
42.
Schadendorf
D
,
Hodi
FS
,
Robert
C
,
Weber
JS
,
Margolin
K
,
Hamid
O
, et al
Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma
.
J Clin Oncol
2015
;
33
:
1889
94
.
43.
Perets
R
,
Bar
J
,
Rasco
DW
,
Ahn
M-J
,
Yoh
K
,
Kim
D-W
, et al
Safety and efficacy of quavonlimab, a novel anti-CTLA-4 antibody (MK-1308), in combination with pembrolizumab in first-line advanced non-small-cell lung cancer
.
Ann Oncol
2020
;
S0923–7534
:
43175
8
.
44.
Korman
AJ
,
Engelhardt
J
,
Loffredo
J
,
Valle
J
,
Akter
R
,
Vuyyuru
R
, et al
Next-generation anti-CTLA-4 antibodies [abstract]
.
In
:
Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1–5
;
Washington, DC. Philadelphia (PA)
:
AACR
; 
2017
.
Abstract nr SY09-01
.
45.
Robert
C
,
Schachter
J
,
Long
GV
,
Arance
A
,
Grob
JJ
,
Mortier
L
, et al
Pembrolizumab versus ipilimumab in advanced melanoma
.
N Engl J Med
2015
;
372
:
2521
32
.
46.
Weber
JS
,
D'Angelo
SP
,
Minor
D
,
Hodi
FS
,
Gutzmer
R
,
Neyns
B
, et al
Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial
.
Lancet Oncol
2015
;
16
:
375
84
.
47.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
48.
Le
DT
,
Durham
JN
,
Smith
KN
,
Wang
H
,
Bartlett
BR
,
Aulakh
LK
, et al
Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade
.
Science
2017
;
357
:
409
13
.
49.
Marabelle
A
,
Fakih
M
,
Lopez
J
,
Shah
M
,
Shapira-Frommer
R
,
Nakagawa
K
, et al
Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study
.
Lancet Oncol
2020
;
21
:
1353
65
.
50.
Antonia
SJ
,
Villegas
A
,
Daniel
D
,
Vicente
D
,
Murakami
S
,
Hui
R
, et al
Durvalumab after chemoradiotherapy in stage III non–small-cell lung cancer
.
N Engl J Med
2017
;
377
:
1919
29
.
51.
Horn
L
,
Mansfield
AS
,
Szcze¸sna
A
,
Havel
L
,
Krzakowski
M
,
Hochmair
MJ
, et al
First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer
.
N Engl J Med
2018
;
379
:
2220
9
.
52.
Paz-Ares
L
,
Dvorkin
M
,
Chen
Y
,
Reinmuth
N
,
Hotta
K
,
Trukhin
D
, et al
Durvalumab plus platinum–etoposide versus platinum–etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): a randomised, controlled, open-label, phase 3 trial
.
Lancet
2019
;
394
:
1929
39
.
53.
Powles
T
,
Park
SH
,
Voog
E
,
Caserta
C
,
Valderrama
BP
,
Gurney
H
, et al
Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma
.
N Engl J Med
2020
;
383
:
1218
30
.
54.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
JJ
,
Cowey
CL
,
Lao
CD
, et al
Combined nivolumab and ipilimumab or monotherapy in untreated melanoma
.
N Engl J Med
2015
;
373
:
23
34
.
55.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
JJ
,
Rutkowski
P
,
Lao
CD
, et al
Five-year survival with combined nivolumab and ipilimumab in advanced melanoma
.
N Engl J Med
2019
;
381
:
1535
46
.
56.
Yau
T
,
Kang
Y-K
,
Kim
T-Y
,
El-Khoueiry
AB
,
Santoro
A
,
Sangro
B
, et al
Efficacy and safety of nivolumab plus ipilimumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib: the CheckMate 040 randomized clinical trial
.
JAMA Oncol
2020
;
6
:
e204564
.
57.
Motzer
RJ
,
Tannir
NM
,
McDermott
DF
,
Arén Frontera
O
,
Melichar
B
,
Choueiri
TK
, et al
Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma
.
N Engl J Med
2018
;
378
:
1277
90
.
58.
Motzer
RJ
,
Escudier
B
,
McDermott
DF
,
Arén Frontera
O
,
Melichar
B
,
Powles
T
, et al
Survival outcomes and independent response assessment with nivolumab plus ipilimumab versus sunitinib in patients with advanced renal cell carcinoma: 42-month follow-up of a randomized phase 3 clinical trial
.
J Immunother Cancer
2020
;
8
:
e000891
.
59.
Hellmann
MD
,
Paz-Ares
L
,
Bernabe Caro
R
,
Zurawski
B
,
Kim
S-W
,
Carcereny Costa
E
, et al
Nivolumab plus ipilimumab in advanced non–small-cell lung cancer
.
N Engl J Med
2019
;
381
:
2020
31
.
60.
Scherpereel
A
,
Mazieres
J
,
Greillier
L
,
Lantuejoul
S
,
P
,
Bylicki
O
, et al
Nivolumab or nivolumab plus ipilimumab in patients with relapsed malignant pleural mesothelioma (IFCT-1501 MAPS2): a multicentre, open-label, randomised, non-comparative, phase 2 trial
.
Lancet Oncol
2019
;
20
:
239
53
.
61.
Overman
MJ
,
Lonardi
S
,
Wong
KYM
,
Lenz
HJ
,
Gelsomino
F
,
Aglietta
M
, et al
Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer
.
J Clin Oncol
2018
;
36
:
773
9
.
62.
Leonetti
A
,
Wever
B
,
Mazzaschi
G
,
Assaraf
YG
,
Rolfo
C
,
Quaini
F
, et al
Molecular basis and rationale for combining immune checkpoint inhibitors with chemotherapy in non-small cell lung cancer
.
Drug Resist Updat
2019
;
46
:
100644
.
63.
Gandhi
L
,
Rodríguez-Abreu
D
,
Gadgeel
S
,
Esteban
E
,
Felip
E
,
De Angelis
F
, et al
Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer
.
N Engl J Med
2018
;
378
:
2078
92
.
64.
Paz-Ares
L
,
Luft
A
,
Vicente
D
,
Tafreshi
A
,
Gümüş
M
,
Mazières
J
, et al
Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer
.
N Engl J Med
2018
;
379
:
2040
51
.
65.
Socinski
MA
,
Jotte
RM
,
Cappuzzo
F
,
Orlandi
F
,
Stroyakovskiy
D
,
Nogami
N
, et al
Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC
.
N Engl J Med
2018
;
378
:
2288
301
.
66.
West
H
,
McCleod
M
,
Hussein
M
,
Morabito
A
,
Rittmeyer
A
,
Conter
HJ
, et al
Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): a multicentre, randomised, open-label, phase 3 tria
.
Lancet Oncol
2019
;
20
:
924
37
.
67.
Schmid
P
,
Adams
S
,
Rugo
HS
,
Schneeweiss
A
,
Barrios
CH
,
Iwata
H
, et al
Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer
.
N Engl J Med
2018
;
379
:
2108
21
.
68.
Burtness
B
,
Harrington
KJ
,
Greil
R
,
Soulières
D
,
Tahara
M
,
de Castro
G
, et al
Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): a randomised, open-label, phase 3 study
.
Lancet
2019
;
394
:
1915
28
.
69.
Makker
V
,
Rasco
D
,
Vogelzang
NJ
,
Brose
MS
,
Cohn
AL
,
Mier
J
, et al
Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer: an interim analysis of a multicentre, open-label, single-arm, phase 2 trial
.
Lancet Oncol
2019
;
20
:
711
8
.
70.
Makker
V
,
Taylor
MH
,
Aghajanian
C
,
Oaknin
A
,
Mier
J
,
Cohn
AL
, et al
Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer
.
J Clin Oncol
2020
;
38
:
2981
92
.
71.
Rini
BI
,
Plimack
ER
,
Stus
V
,
Gafanov
R
,
Hawkins
R
,
Nosov
D
, et al
Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma
.
N Engl J Med
2019
;
380
:
1116
27
.
72.
Plimack
ER
,
Rini
BI
,
Stus
V
,
Gafanov
R
,
Waddell
T
,
Nosov
D
, et al
Pembrolizumab plus axitinib versus sunitinib as first-line therapy for advanced renal cell carcinoma (RCC): updated analysis of KEYNOTE-426
.
J Clin Oncol
2020
;
38
:
5001
.
73.
Motzer
RJ
,
Penkov
K
,
Haanen
J
,
Rini
B
,
Albiges
L
,
Campbell
MT
, et al
Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma
.
N Engl J Med
2019
;
380
:
1103
15
.
74.
Choueiri
TK
,
Powles
T
,
Burotto
M
,
Bourlon
MT
,
Zurawski
B
,
Oyervides Juárez
VM
, et al
696O_PR Nivolumab + cabozantinib vs sunitinib in first-line treatment for advanced renal cell carcinoma: first results from the randomized phase III CheckMate 9ER trial
.
Ann Oncol
2020
;
31
:
S1159
.
75.
Finn
RS
,
Qin
S
,
Ikeda
M
,
Galle
PR
,
Ducreux
M
,
Kim
T-Y
, et al
Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma
.
N Engl J Med
2020
;
382
:
1894
905
.
76.
Gutzmer
R
,
Stroyakovskiy
D
,
Gogas
H
,
Robert
C
,
Lewis
K
,
Protsenko
S
, et al
Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAFV600 mutation-positive melanoma (IMspire150): primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial
.
Lancet
2020
;
395
:
1835
44
.
77.
Brahmer
JR
,
Lacchetti
C
,
Schneider
BJ
,
Atkins
MB
,
Brassil
KJ
,
Caterino
JM
, et al
Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology clinical practice guideline
.
J Clin Oncol
2018
;
36
:
1714
68
.
78.
Abu-Sbeih
H
,
Ali
FS
,
Wang
X
,
Mallepally
N
,
Chen
E
,
Altan
M
, et al
Early introduction of selective immunosuppressive therapy associated with favorable clinical outcomes in patients with immune checkpoint inhibitor-induced colitis
.
J Immunother Cancer
2019
;
7
:
1
11
.
79.
Subudhi
SK
,
Aparicio
A
,
Gao
J
,
Zurita
AJ
,
Araujo
JC
,
Logothetis
CJ
, et al
Clonal expansion of CD8 T cells in the systemic circulation precedes development of ipilimumab-induced toxicities
.
Proc Natl Acad Sci U S A
2016
;
113
:
11919
24
.
80.
Tahir
SA
,
Gao
J
,
Miura
Y
,
Blando
J
,
Tidwell
RSS
,
Zhao
H
, et al
Autoimmune antibodies correlate with immune checkpoint therapy-induced toxicities
.
Proc Natl Acad Sci U S A
2019
;
116
:
22246
51
.
81.
Campbell
MT
,
Zhang
T
,
Chin
L
,
Warner
AB
,
Mathew
M
. 
ApricityRx companion digital therapeutic for evidence-based mitigation and phenotype-linked molecular characterization of irAEs in patients receiving immune checkpoint therapy (ICT)
.
J Clin Oncol
2020
;
38
:
TPS2089
.
82.
Maher
VE
,
Fernandes
LL
,
Weinstock
C
,
Tang
S
,
Agarwal
S
,
Brave
M
, et al
Analysis of the association between adverse events and outcome in patients receiving a programmed death protein 1 or programmed death ligand 1 antibody
.
J Clin Oncol
2019
;
37
:
2730
7
.
83.
Indini
A
,
Di Guardo
L
,
Cimminiello
C
,
Prisciandaro
M
,
Randon
G
,
De Braud
F
, et al
Immune-related adverse events correlate with improved survival in patients undergoing anti-PD1 immunotherapy for metastatic melanoma
.
J Cancer Res Clin Oncol
2019
;
145
:
511
21
.
84.
Sato
K
,
Akamatsu
H
,
Murakami
E
,
Sasaki
S
,
Kanai
K
,
Hayata
A
, et al
Correlation between immune-related adverse events and efficacy in non-small cell lung cancer treated with nivolumab
.
Lung Cancer
2018
;
115
:
71
4
.
85.
Mian
I
,
Yang
M
,
Zhao
H
,
Shah
M
,
Diab
A
,
Shannon
V
, et al
Immune-related adverse events and survival in elderly patients with melanoma treated with ipilimumab
.
J Clin Oncol
2016
;
34
:
3047
.
86.
Owen
DH
,
Wei
L
,
Bertino
EM
,
Edd
T
,
Villalona-Calero
MA
,
He
K
, et al
Incidence, risk factors, and effect on survival of immune-related adverse events in patients with non-small-cell lung cancer
.
Clin Lung Cancer
2018
;
19
:
e893
900
.
87.
Horvat
TZ
,
Adel
NG
,
Dang
T-O
,
Momtaz
P
,
Postow
MA
,
Callahan
MK
, et al
Immune-related adverse events, need for systemic immunosuppression, and effects on survival and time to treatment failure in patients with melanoma treated with ipilimumab at Memorial Sloan Kettering Cancer Center
.
J Clin Oncol
2015
;
33
:
3193
8
.
88.
Ricciuti
B
,
Genova
C
,
De Giglio
A
,
Bassanelli
M
,
Dal Bello
MG
,
Metro
G
, et al
Impact of immune-related adverse events on survival in patients with advanced non-small cell lung cancer treated with nivolumab: long-term outcomes from a multi-institutional analysis
.
J Cancer Res Clin Oncol
2019
;
145
:
479
85
.
89.
Xing
P
,
Zhang
F
,
Wang
G
,
Xu
Y
,
Li
C
,
Wang
S
, et al
Incidence rates of immune-related adverse events and their correlation with response in advanced solid tumours treated with NIVO or NIVO+IPI: a systematic review and meta-analysis
.
J Immunother Cancer
2019
;
7
:
341
.
90.
Das
S
,
Johnson
DB
. 
Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors
.
J Immunother Cancer
2019
;
7
:
306
.
91.
Riudavets
M
,
Mosquera
J
,
Garcia-Campelo
R
,
Serra
J
,
Anguera
G
,
Gallardo
P
, et al
Immune-related adverse events and corticosteroid use for cancer-related symptoms are associated with efficacy in patients with non-small cell lung cancer receiving anti-PD-(L)1 blockade agents
.
Front Oncol
2020
;
10
:
1677
.
92.
Rogado
J
,
Sánchez-Torres
JM
,
Romero-Laorden
N
,
Ballesteros
AI
,
Pacheco-Barcia
V
,
Ramos-Leví
A
, et al
Immune-related adverse events predict the therapeutic efficacy of anti-PD-1 antibodies in cancer patients
.
Eur J Cancer
2019
;
109
:
21
7
.
93.
Toi
Y
,
Sugawara
S
,
Kawashima
Y
,
Aiba
T
,
Kawana
S
,
Saito
R
, et al
Association of immune-related adverse events with clinical benefit in patients with advanced non-small-cell lung cancer treated with nivolumab
.
Oncologist
2018
;
23
:
1358
65
.
94.
Okada
N
,
Kawazoe
H
,
Takechi
K
,
Matsudate
Y
,
Utsunomiya
R
,
Zamami
Y
, et al
Association between immune-related adverse events and clinical efficacy in patients with melanoma treated with nivolumab: a multicenter retrospective study
.
Clin Ther
2019
;
41
:
59
67
.
95.
Elias
R
,
Yan
F
,
Singla
N
,
Levonyack
N
,
Formella
J
,
Christie
A
, et al
Immune-related adverse events are associated with improved outcomes in ICI-treated renal cell carcinoma patients
.
J Clin Oncol
2019
;
37
:
645
.
96.
Grangeon
M
,
Tomasini
P
,
Chaleat
S
,
Jeanson
A
,
Souquet-Bressand
M
,
Khobta
N
, et al
Association between immune-related adverse events and efficacy of immune checkpoint inhibitors in non-small-cell lung cancer
.
Clin Lung Cancer
2019
;
20
:
201
7
.
97.
Wei
SC
,
Meijers
WC
,
Axelrod
ML
,
Anang
N-AAS
,
Screever
EM
,
Wescott
EC
, et al
A genetic mouse model recapitulates immune checkpoint inhibitor-associated myocarditis and supports a mechanism-based therapeutic intervention
.
Cancer Discov
2021
;
11
:
1
12
.
98.
Salem
J-E
,
Allenbach
Y
,
Vozy
A
,
Brechot
N
,
Johnson
DB
,
Moslehi
JJ
, et al
Abatacept for severe immune checkpoint inhibitor–associated myocarditis
.
N Engl J Med
2019
;
380
:
2377
9
.
99.
Luoma
AM
,
Suo
S
,
Williams
HL
,
Sharova
T
,
Sullivan
K
,
Manos
M
, et al
Molecular pathways of colon inflammation induced by cancer immunotherapy
.
Cell
2020
;
182
:
655
71
.
100.
Kwon
ED
,
Drake
CG
,
Scher
HI
,
Fizazi
K
,
Bossi
A
,
van den Eertwegh
AJM
, et al
Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial
.
Lancet Oncol
2014
;
15
:
700
12
.
101.
Beer
TM
,
Kwon
ED
,
Drake
CG
,
Fizazi
K
,
Logothetis
C
,
Gravis
G
, et al
Randomized, double-blind, phase III trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic chemotherapy-naive castration-resistant prostate cancer
.
J Clin Oncol
2017
;
35
:
40
7
.
102.
Antonarakis
ES
,
Piulats
JM
,
Gross-Goupil
M
,
Goh
J
,
Ojamaa
K
,
Hoimes
CJ
, et al
Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: multicohort, open-label phase II KEYNOTE-199 study
.
J Clin Oncol
2020
;
38
:
395
405
.
103.
Sweeney
CJ
,
Gillessen
S
,
Rathkopf
D
,
Matsubara
N
,
Drake
C
,
Fizazi
K
, et al
Abstract CT014: IMbassador250: a phase III trial comparing atezolizumab with enzalutamide vs enzalutamide alone in patients with metastatic castration-resistant prostate cancer (mCRPC)
.
Cancer Res
2020
;
CT014 LP–CT014
.
104.
Antonarakis
ES
,
Goh
JC
,
Gross-Goupil
M
,
Vaishampayan
UN
,
Piulats
JM
,
De Wit
R
, et al
Pembrolizumab for metastatic castration-resistant prostate cancer (mCRPC) previously treated with docetaxel: updated analysis of KEYNOTE-199
.
J Clin Oncol
2019
;
37
:
216
.
105.
Gao
J
,
Ward
JF
,
Pettaway
CA
,
Shi
LZ
,
Subudhi
SK
,
Vence
LM
, et al
VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer
.
Nat Med
2017
;
23
:
551
5
.
106.
Bishop
JL
,
Sio
A
,
Angeles
A
,
Roberts
ME
,
Azad
AA
,
Chi
KN
, et al
PD-L1 is highly expressed in enzalutamide resistant prostate cancer
.
Oncotarget
2015
;
6
:
234
42
.
107.
Fizazi
K
,
Drake
CG
,
Beer
TM
,
Kwon
ED
,
Scher
HI
,
Gerritsen
WR
, et al
Final analysis of the ipilimumab versus placebo following radiotherapy phase III trial in postdocetaxel metastatic castration-resistant prostate cancer identifies an excess of long-term survivors
.
Eur Urol
2020
;
78
:
822
30
.
108.
Lines
JL
,
Pantazi
E
,
Mak
J
,
Sempere
LF
,
Wang
L
,
O'Connell
S
, et al
VISTA is an immune checkpoint molecule for human T cells
.
Cancer Res
2014
;
74
:
1924
32
.
109.
Sharma
P
,
Pachynski
RK
,
Narayan
V
,
Fléchon
A
,
Gravis
G
,
Galsky
MD
, et al
Nivolumab plus ipilimumab for metastatic castration-resistant prostate cancer: preliminary analysis of patients in the CheckMate 650 trial
.
Cancer Cell
2020
;
38
:
489
99
.
110.
Jiao
S
,
Subudhi
SK
,
Aparicio
A
,
Ge
Z
,
Guan
B
,
Miura
Y
, et al
Differences in tumor microenvironment dictate T helper lineage polarization and response to immune checkpoint therapy
.
Cell
2019
;
179
:
1177
90
.
111.
Lee
JC
,
Mehdizadeh
S
,
Smith
J
,
Young
A
,
Mufazalov
IA
,
Mowery
CT
, et al
Regulatory T cell control of systemic immunity and immunotherapy response in liver metastasis
.
Sci Immunol
2020
;
5
:
eaba0759
.
112.
Goswami
S
,
Walle
T
,
Cornish
AE
,
Basu
S
,
Anandhan
S
,
Fernandez
I
, et al
Immune profiling of human tumors identifies CD73 as a combinatorial target in glioblastoma
.
Nat Med
2020
;
26
:
39
46
.
113.
Najafi
M
,
Hashemi Goradel
N
,
Farhood
B
,
Salehi
E
,
Nashtaei
MS
,
Khanlarkhani
N
, et al
Macrophage polarity in cancer: a review
.
J Cell Biochem
2019
;
120
:
2756
65
.
114.
Asare
PF
,
Roscioli
E
,
Hurtado
PR
,
Tran
HB
,
Mah
CY
,
Hodge
S
. 
LC3-associated phagocytosis (LAP): a potentially influential mediator of efferocytosis-related tumor progression and aggressiveness
.
Front Oncol
2020
;
10
:
1298
.
115.
Flavahan
WA
,
Gaskell
E
,
Bernstein
BE
. 
Epigenetic plasticity and the hallmarks of cancer
.
Science
2017
;
357
:
eaal2380
.
116.
Goswami
S
,
Apostolou
I
,
Zhang
J
,
Skepner
J
,
Anandhan
S
,
Zhang
X
, et al
Modulation of EZH2 expression in T cells improves efficacy of anti-CTLA-4 therapy
.
J Clin Invest
2018
;
128
:
3813
8
.
117.
Suvà
ML
,
Riggi
N
,
Bernstein
BE
. 
Epigenetic reprogramming in cancer
.
Science
2013
;
339
:
1567
70
.
118.
Kim
KH
,
Roberts
CWM
. 
Targeting EZH2 in cancer
.
Nat Med
2016
;
22
:
128
34
.
119.
DuPage
M
,
Chopra
G
,
Quiros
J
,
Rosenthal
WL
,
Morar
MM
,
Holohan
D
, et al
The chromatin-modifying enzyme Ezh2 is critical for the maintenance of regulatory T cell identity after activation
.
Immunity
2015
;
42
:
227
38
.
120.
Villanueva
L
,
Álvarez-Errico
D
,
Esteller
M
. 
The contribution of epigenetics to cancer immunotherapy
.
Trends Immunol
2020
;
41
:
676
91
.
121.
Sharma
P
,
Allison
JP
. 
Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential
.
Cell
2015
;
161
:
205
14
.
122.
Hamid
O
,
Schmidt
H
,
Nissan
A
,
Ridolfi
L
,
Aamdal
S
,
Hansson
J
, et al
A prospective phase II trial exploring the association between tumor microenvironment biomarkers and clinical activity of ipilimumab in advanced melanoma
.
J Transl Med
2011
;
9
:
204
.
123.
Spranger
S
,
Bao
R
,
Gajewski
TF
. 
Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity
.
Nature
2015
;
523
:
231
5
.
124.
Peng
W
,
Chen
JQ
,
Liu
C
,
Malu
S
,
Creasy
C
,
Tetzlaff
MT
, et al
Loss of PTEN promotes resistance to T cell-mediated immunotherapy
.
Cancer Discov
2016
;
6
:
202
16
.
125.
Snyder
A
,
Makarov
V
,
Merghoub
T
,
Yuan
J
,
Zaretsky
JM
,
Desrichard
A
, et al
Genetic basis for clinical response to CTLA-4 blockade in melanoma
.
N Engl J Med
2014
;
371
:
2189
99
.
126.
Hellmann
MD
,
Nathanson
T
,
Rizvi
H
,
Creelan
BC
,
Sanchez-Vega
F
,
Ahuja
A
, et al
Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer
.
Cancer Cell
2018
;
33
:
843
52
.
127.
Ganesan
S
,
Mehnert
J
. 
Biomarkers for response to immune checkpoint blockade
.
Annu Rev Cancer Biol
2020
;
4
:
331
51
.
128.
Yarchoan
M
,
Hopkins
A
,
Jaffee
EM
. 
Tumor mutational burden and response rate to PD-1 inhibition
.
N Engl J Med
2017
;
377
:
2500
1
.
129.
Braun
DA
,
Bakouny
Z
,
Hirsch
L
,
Flippot
R
,
Van Allen
EM
,
Wu
CJ
, et al
Beyond conventional immune-checkpoint inhibition — novel immunotherapies for renal cell carcinoma
.
Nat Rev Clin Oncol
2021
Jan 12 [Epub ahead of print]
.
130.
Cristescu
R
,
Mogg
R
,
Ayers
M
,
Albright
A
,
Murphy
E
,
Yearley
J
, et al
Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy
.
Science
2018
;
362
:
eaar3593
.
131.
Subudhi
SK
,
Vence
L
,
Zhao
H
,
Blando
J
,
Yadav
SS
,
Xiong
Q
, et al
Neoantigen responses, immune correlates, and favorable outcomes after ipilimumab treatment of patients with prostate cancer
.
Sci Transl Med
2020
;
12
:
1
11
.
132.
Braun
DA
,
Hou
Y
,
Bakouny
Z
,
Ficial
M
,
Sant' Angelo
M
,
Forman
J
, et al
Interplay of somatic alterations and immune infiltration modulates response to PD-1 blockade in advanced clear cell renal cell carcinoma
.
Nat Med
2020
;
26
:
909
18
.
133.
Ott
PA
,
Hu-Lieskovan
S
,
Chmielowski
B
,
Govindan
R
,
Naing
A
,
Bhardwaj
N
, et al
A phase Ib trial of personalized neoantigen therapy plus anti-PD-1 in patients with advanced melanoma, non-small cell lung cancer, or bladder cancer
.
Cell
2020
;
183
:
347
62
.
134.
Keskin
DB
,
Anandappa
AJ
,
Sun
J
,
Tirosh
I
,
Mathewson
ND
,
Li
S
, et al
Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial
.
Nature
2019
;
565
:
234
9
.
135.
Ott
PA
,
Hu
Z
,
Keskin
DB
,
Shukla
SA
,
Sun
J
,
Bozym
DJ
, et al
An immunogenic personal neoantigen vaccine for patients with melanoma
.
Nature
2017
;
547
:
217
21
.
136.
Stewart
RA
,
Pilie
PG
,
Yap
TA
. 
Development of PARP and immune-checkpoint inhibitor combinations
.
Cancer Res
2018
;
78
:
6717
25
.
137.
Sen
T
,
Rodriguez
BL
,
Chen
L
,
Della Corte
CM
,
Morikawa
N
,
Fujimoto
J
, et al
Targeting DNA damage response promotes antitumor immunity through STING-mediated T-cell activation in small cell lung cancer
.
Cancer Discov
2019
;
9
:
646
61
.
138.
Parkes
EE
,
Walker
SM
,
Taggart
LE
,
McCabe
N
,
Knight
LA
,
Wilkinson
R
, et al
Activation of STING-dependent innate immune signaling by s-phase-specific DNA damage in breast cancer
.
J Natl Cancer Inst
2017
;
109
:
1
10
.
139.
Härtlova
A
,
Erttmann
SF
,
Raffi
FA
,
Schmalz
AM
,
Resch
U
,
Anugula
S
, et al
DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity
.
Immunity
2015
;
42
:
332
43
.
140.
Teo
MY
,
Seier
K
,
Ostrovnaya
I
,
Regazzi
AM
,
Kania
BE
,
Moran
MM
, et al
Alterations in DNA damage response and repair genes as potential marker of clinical benefit from PD-1/PD-L1 blockade in advanced urothelial cancers
.
J Clin Oncol
2018
;
36
:
1685
94
.
141.
Samstein
RM
,
Krishna
C
,
Ma
X
,
Pei
X
,
Lee
K-W
,
Makarov
V
, et al
Mutations in BRCA1 and BRCA2 differentially affect the tumor microenvironment and response to checkpoint blockade immunotherapy
.
Nat Cancer
2020
;
1
:
1188
203
.
142.
Miao
D
,
Margolis
CA
,
Vokes
NI
,
Liu
D
,
Taylor-Weiner
A
,
Wankowicz
SM
, et al
Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors
.
Nat Genet
2018
;
50
:
1271
81
.
143.
Rosenberg
JE
,
Hoffman-Censits
J
,
Powles
T
,
Van Der Heijden
MS
,
Balar
AV
,
Necchi
A
, et al
Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial
.
Lancet
2016
;
387
:
1909
20
.
144.
Balar
AV
,
Galsky
MD
,
Rosenberg
JE
,
Powles
T
,
Petrylak
DP
,
Bellmunt
J
, et al
Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial
.
Lancet
2017
;
389
:
67
76
.
145.
Rosenberg
SA
,
Packard
BS
,
Aebersold
PM
,
Solomon
D
,
Topalian
SL
,
Toy
ST
, et al
Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report
.
N Engl J Med
1988
;
319
:
1676
80
.
146.
Zhang
L
,
Conejo-Garcia
JR
,
Katsaros
D
,
Gimotty
PA
,
Massobrio
M
,
Regnani
G
, et al
Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer
.
N Engl J Med
2003
;
348
:
203
13
.
147.
Sharma
P
,
Shen
Y
,
Wen
S
,
Yamada
S
,
Jungbluth
AA
,
Gnjatic
S
, et al
CD8 tumor-infiltrating lymphocytes are predictive of survival in muscle-invasive urothelial carcinoma
.
Proc Natl Acad Sci U S A
2007
;
104
:
3967
72
.
148.
Hodi
FS
,
Butler
M
,
Oble
DA
,
Seiden
MV
,
Haluska
FG
,
Kruse
A
, et al
Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients
.
Proc Natl Acad Sci U S A
2008
;
105
:
3005
10
.
149.
Tumeh
PC
,
Harview
CL
,
Yearley
JH
,
Shintaku
IP
,
Taylor
EJM
,
Robert
L
, et al
PD-1 blockade induces responses by inhibiting adaptive immune resistance
.
Nature
2014
;
515
:
568
71
.
150.
Loi
S
,
Drubay
D
,
Adams
S
,
Pruneri
G
,
Francis
PA
,
Lacroix-Triki
M
, et al
Tumor-infiltrating lymphocytes and prognosis: a pooled individual patient analysis of early-stage triple-negative breast cancers
.
J Clin Oncol
2019
;
37
:
559
69
.
151.
Galon
J
,
Fox
BA
,
Bifulco
CB
,
Masucci
G
,
Rau
T
,
Botti
G
, et al
Immunoscore and immunoprofiling in cancer: an update from the melanoma and immunotherapy bridge 2015
.
J Transl Med
2016
;
14
:
273
.
152.
Bindea
G
,
Mlecnik
B
,
Angell
HK
,
Galon
J
. 
The immune landscape of human tumors: implications for cancer immunotherapy
.
Oncoimmunology
2014
;
3
:
e27456
.
153.
Sade-Feldman
M
,
Yizhak
K
,
Bjorgaard
SL
,
Ray
JP
,
de Boer
CG
,
Jenkins
RW
, et al
Defining T cell states associated with response to checkpoint immunotherapy in melanoma
.
Cell
2018
;
175
:
998
1013
.
154.
Balatoni
T
,
Mohos
A
,
Papp
E
,
Sebestyén
T
,
Liszkay
G
,
Oláh
J
, et al
Tumor-infiltrating immune cells as potential biomarkers predicting response to treatment and survival in patients with metastatic melanoma receiving ipilimumab therapy
.
Cancer Immunol Immunother
2018
;
67
:
141
51
.
155.
Luke
JJ
,
Bao
R
,
Sweis
RF
,
Spranger
S
,
Gajewski
TF
. 
WNT/β-catenin pathway activation correlates with immune exclusion across human cancers
.
Clin Cancer Res
2019
;
25
:
3074
83
.
156.
Trujillo
JA
,
Luke
JJ
,
Zha
Y
,
Segal
JP
,
Ritterhouse
LL
,
Spranger
S
, et al
Secondary resistance to immunotherapy associated with β-catenin pathway activation or PTEN loss in metastatic melanoma
.
J Immunother Cancer
2019
;
7
:
295
.
157.
Dighe
AS
,
Richards
E
,
Old
LJ
,
Schreiber
RD
. 
Enhanced in vivo growth and resistance to rejection of tumor cells expressing dominant negative IFN gamma receptors
.
Immunity
1994
;
1
:
447
56
.
158.
Kaplan
DH
,
Shankaran
V
,
Dighe
AS
,
Stockert
E
,
Aguet
M
,
Old
LJ
, et al
Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice
.
Proc Natl Acad Sci U S A
1998
;
95
:
7556
61
.
159.
Gao
J
,
Shi
LZ
,
Zhao
H
,
Chen
J
,
Xiong
L
,
He
Q
, et al
Loss of IFNγ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy
.
Cell
2016
;
167
:
397
404
.
160.
Zaretsky
JM
,
Garcia-Diaz
A
,
Shin
DS
,
Escuin-Ordinas
H
,
Hugo
W
,
Hu-Lieskovan
S
, et al
Mutations associated with acquired resistance to PD-1 blockade in melanoma
.
N Engl J Med
2016
;
375
:
819
29
.
161.
Ayers
M
,
Lunceford
J
,
Nebozhyn
M
,
Murphy
E
,
Loboda
A
,
Kaufman
DR
, et al
IFNγ–related mRNA profile predicts clinical response to PD-1 blockade
.
J Clin Invest
2017
;
127
:
2930
40
.
162.
Grasso
CS
,
Tsoi
J
,
Onyshchenko
M
,
Abril-Rodriguez
G
,
Ross-Macdonald
P
,
Wind-Rotolo
M
, et al
Conserved interferon-γ signaling drives clinical response to immune checkpoint blockade therapy in melanoma
.
Cancer Cell
2020
;
38
:
500
15
.
163.
Petitprez
F
,
de Reyniès
A
,
Keung
EZ
,
Chen
TW-W
,
Sun
C-M
,
Calderaro
J
, et al
B cells are associated with survival and immunotherapy response in sarcoma
.
Nature
2020
;
577
:
556
60
.
164.
Helmink
BA
,
Reddy
SM
,
Gao
J
,
Zhang
S
,
Basar
R
,
Thakur
R
, et al
B cells and tertiary lymphoid structures promote immunotherapy response
.
Nature
2020
;
577
:
549
55
.
165.
Koti
M
,
Xu
AS
,
Ren
KYM
,
Visram
K
,
Ren
R
,
Berman
DM
, et al
Tertiary lymphoid structures associate with tumour stage in urothelial bladder cancer
.
Bladder Cancer
2017
;
3
:
259
67
.
166.
Germain
C
,
Gnjatic
S
,
Tamzalit
F
,
Knockaert
S
,
Remark
R
,
Goc
J
, et al
Presence of B cells in tertiary lymphoid structures is associated with a protective immunity in patients with lung cancer
.
Am J Respir Crit Care Med
2014
;
189
:
832
44
.
167.
Thommen
DS
,
Koelzer
VH
,
Herzig
P
,
Roller
A
,
Trefny
M
,
Dimeloe
S
, et al
A transcriptionally and functionally distinct PD-1(+) CD8(+) T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade
.
Nat Med
2018
;
24
:
994
1004
.
168.
Sautès-Fridman
C
,
Petitprez
F
,
Calderaro
J
,
Fridman
WH
. 
Tertiary lymphoid structures in the era of cancer immunotherapy
.
Nat Rev Cancer
2019
;
19
:
307
25
.
169.
Litchfield
K
,
Reading
JL
,
Puttick
C
,
Thakkar
K
,
Abbosh
C
,
Bentham
R
, et al
Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition
.
Cell
2021
;
184
:
596
614
.
170.
Goswami
S
,
Chen
Y
,
Anandhan
S
,
Szabo
PM
,
Basu
S
,
Blando
JM
, et al
ARID1A mutation plus CXCL13 expression act as combinatorial biomarkers to predict responses to immune checkpoint therapy in mUCC
.
Sci Transl Med
2020
;
12
:
eabc4220
.
171.
Fu
T
,
He
Q
,
Sharma
P
. 
The ICOS/ICOSL pathway is required for optimal antitumor responses mediated by anti-CTLA-4 therapy
.
Cancer Res
2011
;
71
:
5445
54
.
172.
Fan
X
,
Quezada
SA
,
Sepulveda
MA
,
Sharma
P
,
Allison
JP
. 
Engagement of the ICOS pathway markedly enhances efficacy of CTLA-4 blockade in cancer immunotherapy
.
J Exp Med
2014
;
211
:
715
25
.
173.
Amaria
RN
,
Reddy
SM
,
Tawbi
HA
,
Davies
MA
,
Ross
MI
,
Glitza
IC
, et al
Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma
.
Nat Med
2018
;
24
:
1649
54
.
174.
Rozeman
EA
,
Menzies
AM
,
van Akkooi
ACJ
,
Adhikari
C
,
Bierman
C
,
van de Wiel
BA
, et al
Identification of the optimal combination dosing schedule of neoadjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma (OpACIN-neo): a multicentre, phase 2, randomised, controlled trial
.
Lancet Oncol
2019
;
20
:
948
60
.
175.
Nghiem
P
,
Bhatia
S
,
Lipson
EJ
,
Sharfman
WH
,
Kudchadkar
RR
,
Brohl
AS
, et al
Durable tumor regression and overall survival in patients with advanced Merkel cell carcinoma receiving pembrolizumab as first-line therapy
.
J Clin Oncol
2019
;
37
:
693
702
.
176.
Forde
PM
,
Chaft
JE
,
Smith
KN
,
Anagnostou
V
,
Cottrell
TR
,
Hellmann
MD
, et al
Neoadjuvant PD-1 blockade in resectable lung cancer
.
N Engl J Med
2018
;
378
:
1976
86
.
177.
Chalabi
M
,
Fanchi
LF
,
Dijkstra
KK
,
Van den Berg
JG
,
Aalbers
AG
,
Sikorska
K
, et al
Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers
.
Nat Med
2020
;
26
:
566
76
.
178.
Necchi
A
,
Anichini
A
,
Raggi
D
,
Briganti
A
,
Massa
S
,
Lucianò
R
, et al
Pembrolizumab as neoadjuvant therapy before radical cystectomy in patients with muscle-invasive urothelial bladder carcinoma (PURE-01): an open-label, single-arm, phase II study
.
J Clin Oncol
2018
;
36
:
3353
60
.
179.
Topalian
SL
,
Bhatia
S
,
Amin
A
,
Kudchadkar
RR
,
Sharfman
WH
,
Lebbé
C
, et al
Neoadjuvant nivolumab for patients with resectable merkel cell carcinoma in the CheckMate 358 trial
.
J Clin Oncol
2020
;
38
:
2476
87
.
180.
Nanda
R
,
Liu
MC
,
Yau
C
,
Shatsky
R
,
Pusztai
L
,
Wallace
A
, et al
Effect of pembrolizumab plus neoadjuvant chemotherapy on pathologic complete response in women with early-stage breast cancer: an analysis of the ongoing phase 2 adaptively randomized I-SPY2 trial
.
JAMA Oncol
2020
;
6
:
676
84
.
181.
Mittendorf
EA
,
Zhang
H
,
Barrios
CH
,
Saji
S
,
Jung
KH
,
Hegg
R
, et al
Neoadjuvant atezolizumab in combination with sequential nab-paclitaxel and anthracycline-based chemotherapy versus placebo and chemotherapy in patients with early-stage triple-negative breast cancer (IMpassion031): a randomised, double-blind, phase 3 trial
.
Lancet
2020
;
396
:
1090
100
.
182.
Tolaney
SM
,
Jerusalem
G
,
Salgado
R
,
Liu
X
,
Chen
T
,
Zhang
H
, et al
A phase II trial of nivolumab (NIVO) + palbociclib (PAL) + anastrozole (ANA) in postmenopausal women and men with estrogen receptor (ER)+/human epidermal growth factor 2 (HER2)- primary breast cancer (BC): CheckMate 7A8
.
J Clin Oncol
2020
;
38
:
TPS1105
.
183.
Loi
S
,
McArthur
HL
,
Harbeck
N
,
Pusztai
L
,
Delaloge
S
,
Letrent
K
, et al
A phase III trial of nivolumab with neoadjuvant chemotherapy and adjuvant endocrine therapy in ER+/HER2 primary breast cancer: CheckMate 7FL
.
J Clin Oncol
2020
;
38
:
TPS604
.
184.
Cardoso
F
,
Bardia
A
,
Andre
F
,
Cescon
DW
,
McArthur
HL
,
Telli
ML
, et al
KEYNOTE-756: randomized, double-blind, phase 3 study of pembrolizumab vs placebo combined with neoadjuvant chemotherapy and adjuvant endocrine therapy for high-risk, early-stage estrogen receptor–positive, human epidermal growth factor receptor 2–negative
.
J Clin Oncol
2019
;
37
:
TPS601
.
185.
Heymach
J
,
Taube
J
,
Mitsudomi
T
,
Harpole
D
,
Aperghis
M
,
Trani
L
, et al
P1.18-02 the AEGEAN phase 3 trial of neoadjuvant/adjuvant durvalumab in patients with resectable stage II/III NSCLC
.
J Thorac Oncol
2019
;
14
:
S625
6
.
186.
Fernando
HC
,
Yang
J
,
Ferraro
GL
,
Keller
SM
. 
Randomized, double-blind phase 3 study evaluating neoadjuvant platinum-based chemotherapy with perioperative pembrolizumab or placebo in resectable stage IIB or IIIA NSCLC: KEYNOTE-671
.
J Clin Oncol
2018
;
36
:
TPS8583
.
187.
Forde
PM
,
Chaft
JE
,
Felip
E
,
Broderick
S
,
Girard
N
,
Awad
MM
, et al
Checkmate 816: a phase 3, randomized, open-label trial of nivolumab plus ipilimumab vs platinum-doublet chemotherapy as neoadjuvant treatment for early-stage NSCLC
.
J Clin Oncol
2017
;
35
:
TPS8577
.
188.
Sonpavde
G
,
Necchi
A
,
Gupta
S
,
Steinberg
GD
,
Gschwend
JE
,
Van Der Heijden
MS
, et al
A phase 3 randomized study of neoadjuvant chemotherapy (NAC) alone or in combination with nivolumab (NIVO) ± BMS-986205 in cisplatin-eligible muscle invasive bladder cancer (MIBC)
.
J Clin Oncol
2019
;
37
:
TPS4587
.
189.
Eggermont
AMM
,
Blank
CU
,
Mandala
M
,
Long
GV
,
Atkinson
V
,
Dalle
S
, et al
Adjuvant pembrolizumab versus placebo in resected stage III melanoma
.
N Engl J Med
2018
;
378
:
1789
801
.
190.
Eggermont
AMM
,
Chiarion-Sileni
V
,
Grob
J-J
,
Dummer
R
,
Wolchok
JD
,
Schmidt
H
, et al
Adjuvant ipilimumab versus placebo after complete resection of high-risk stage III melanoma (EORTC 18071): a randomised, double-blind, phase 3 trial
.
Lancet Oncol
2015
;
16
:
522
30
.
191.
Weber
J
,
Mandala
M
,
Del Vecchio
M
,
Gogas
HJ
,
Arance
AM
,
Cowey
CL
, et al
Adjuvant nivolumab versus ipilimumab in resected stage III or IV melanoma
.
N Engl J Med
2017
;
377
:
1824
35
.
192.
Tarhini
AA
,
Lee
SJ
,
Hodi
FS
,
Rao
UNM
,
Cohen
GI
,
Hamid
O
, et al
Phase III study of adjuvant ipilimumab (3 or 10 mg/kg) versus high-dose interferon alfa-2b for resected high-risk melanoma: North American Intergroup E1609
.
J Clin Oncol
2020
;
38
:
567
75
.
193.
Ascierto
PA
,
Del Vecchio
M
,
Mandalá
M
,
Gogas
H
,
Arance
AM
,
Dalle
S
, et al
Adjuvant nivolumab versus ipilimumab in resected stage IIIB-C and stage IV melanoma (CheckMate 238): 4-year results from a multicentre, double-blind, randomised, controlled, phase 3 trial
.
Lancet Oncol
2020
;
21
:
1465
77
.
194.
Roudi
R
,
Syn
NL
,
Roudbary
M
. 
Antimicrobial peptides as biologic and immunotherapeutic agents against cancer: a comprehensive overview
.
Front Immunol
2017
;
8
:
1320
.
195.
Bommareddy
PK
,
Shettigar
M
,
Kaufman
HL
. 
Integrating oncolytic viruses in combination cancer immunotherapy
.
Nat Rev Immunol
2018
;
18
:
498
513
.
196.
Zamarin
D
,
Holmgaard
RB
,
Subudhi
SK
,
Park
JS
,
Mansour
M
,
Palese
P
, et al
Localized oncolytic virotherapy overcomes systemic tumor resistance to immune checkpoint blockade immunotherapy
.
Sci Transl Med
2014
;
6
:
226ra32
.
197.
Spranger
S
,
Dai
D
,
Horton
B
,
Gajewski
TF
. 
Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy
.
Cancer Cell
2017
;
31
:
711
23
.
198.
Theisen
DJ
,
Ferris
ST
,
Briseño
CG
,
Kretzer
N
,
Iwata
A
,
Murphy
KM
, et al
Batf3-dependent genes control tumor rejection induced by dendritic cells independently of cross-presentation
.
Cancer Immunol Res
2019
;
7
:
29
39
.
199.
Le Naour
J
,
Zitvogel
L
,
Galluzzi
L
,
Vacchelli
E
,
Kroemer
G
. 
Trial watch: STING agonists in cancer therapy
.
Oncoimmunology
2020
;
9
:
1777624
.
200.
Le Naour
J
,
Galluzzi
L
,
Zitvogel
L
,
Kroemer
G
,
Vacchelli
E
. 
Trial watch: TLR3 agonists in cancer therapy
.
Oncoimmunology
2020
;
9
:
1771143
.
201.
Frega
G
,
Wu
Q
,
Le Naour
J
,
Vacchelli
E
,
Galluzzi
L
,
Kroemer
G
, et al
Trial Watch: experimental TLR7/TLR8 agonists for oncological indications
.
Oncoimmunology
2020
;
9
:
1796002
.
202.
De Keersmaecker
B
,
Claerhout
S
,
Carrasco
J
,
Bar
I
,
Corthals
J
,
Wilgenhof
S
, et al
TriMix and tumor antigen mRNA electroporated dendritic cell vaccination plus ipilimumab: link between T-cell activation and clinical responses in advanced melanoma
.
J Immunother Cancer
2020
;
8
:
e000329
.
203.
Bommareddy
PK
,
Patel
A
,
Hossain
S
,
Kaufman
HL
. 
Talimogene laherparepvec (T-VEC) and other oncolytic viruses for the treatment of melanoma
.
Am J Clin Dermatol
2017
;
18
:
1
15
.
204.
Andtbacka
RHI
,
Collichio
F
,
Harrington
KJ
,
Middleton
MR
,
Downey
G
,
Öhrling
K
, et al
Final analyses of OPTiM: a randomized phase III trial of talimogene laherparepvec versus granulocyte-macrophage colony-stimulating factor in unresectable stage III-IV melanoma
.
J Immunother Cancer
2019
;
7
:
1
11
.
205.
Matson
V
,
Fessler
J
,
Bao
R
,
Chongsuwat
T
,
Zha
Y
,
Alegre
ML
, et al
The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients
.
Science
2018
;
359
:
104
8
.
206.
Gopalakrishnan
V
,
Spencer
CN
,
Nezi
L
,
Reuben
A
,
Andrews
MC
,
Karpinets
TV
, et al
Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients
.
Science
2018
;
359
:
97
103
.
207.
Suntharalingam
G
,
Perry
MR
,
Ward
S
,
Brett
SJ
,
Castello-Cortes
A
,
Brunner
MD
, et al
Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412
.
N Engl J Med
2006
;
355
:
1018
28
.
208.
Patel
MR
,
Ellerton
J
,
Infante
JR
,
Agrawal
M
,
Gordon
M
,
Aljumaily
R
, et al
Avelumab in metastatic urothelial carcinoma after platinum failure (JAVELIN Solid Tumor): pooled results from two expansion cohorts of an open-label, phase 1 trial
.
Lancet Oncol
2018
;
19
:
51
64
.
209.
Chester
C
,
Sanmamed
MF
,
Wang
J
,
Melero
I
. 
Immunotherapy targeting 4-1BB: mechanistic rationale, clinical results, and future strategies
.
Blood
2018
;
131
:
49
57
.
210.
Choi
Y
,
Shi
Y
,
Haymaker
CL
,
Naing
A
,
Ciliberto
G
,
Hajjar
J
. 
T-cell agonists in cancer immunotherapy
.
J Immunother Cancer
2020
;
8
:
e000966
.
211.
Segal
NH
,
Logan
TF
,
Hodi
FS
,
McDermott
D
,
Melero
I
,
Hamid
O
, et al
Results from an integrated safety analysis of urelumab, an agonist anti-CD137 monoclonal antibody
.
Clin Cancer Res
2017
;
23
:
1929
36
.
212.
Segal
NH
,
He
AR
,
Doi
T
,
Levy
R
,
Bhatia
S
,
Pishvaian
MJ
, et al
Phase i study of single-agent utomilumab (PF-05082566), a 4-1bb/cd137 agonist, in patients with advanced cancer
.
Clin Cancer Res
2018
;
24
:
1816
23
.
213.
Rischin
D
,
Groenland
SL
,
Lim
AML
,
Martin-Liberal
J
,
Moreno
V
,
Trigo Perez
JM
, et al
Inducible T cell costimulatory (ICOS) receptor agonist, GSK3359609 (GSK609) alone and in combination with pembrolizumab (Pembro): preliminary results from INDUCE-1 expansion cohorts (EC) in head and neck squamous cell carcinoma (HNSCC)
.
Ann Oncol
2019
;
30
:
v449
74
.
214.
Mayes
PA
,
Hance
KW
,
Hoos
A
. 
The promise and challenges of immune agonist antibody development in cancer
.
Nat Rev Drug Discov
2018
;
17
:
509
27
.
215.
Waite
JC
,
Wang
B
,
Haber
L
,
Hermann
A
,
Ullman
E
,
Ye
X
, et al
Tumor-targeted CD28 bispecific antibodies enhance the antitumor efficacy of PD-1 immunotherapy
.
Sci Transl Med
2020
;
12
:
eaba2325
.
216.
Zhang
Q
,
Luo
J
,
Wu
S
,
Si
H
,
Gao
C
,
Xu
W
, et al
Prognostic and predictive impact of circulating tumor DNA in patients with advanced cancers treated with immune checkpoint blockade
.
Cancer Discov
2020
;
10
:
1842
53
.
217.
Wells
DK
,
van Buuren
MM
,
Dang
KK
,
Hubbard-Lucey
VM
,
Sheehan
KCF
,
Campbell
KM
, et al
Key parameters of tumor epitope immunogenicity revealed through a consortium approach improve neoantigen prediction
.
Cell
2020
;
183
:
818
34
.
218.
Alspach
E
,
Lussier
DM
,
Miceli
AP
,
Kizhvatov
I
,
DuPage
M
,
Luoma
AM
, et al
MHC-II neoantigens shape tumour immunity and response to immunotherapy
.
Nature
2019
;
574
:
696
701
.