Harnessing the immune system to treat cancer through inhibitors of CTLA4 and PD-L1 has revolutionized the landscape of cancer. Rational combination strategies aim to enhance the antitumor effects of immunotherapies, but require a deep understanding of the mechanistic underpinnings of the immune system and robust preclinical and clinical drug development strategies. We review the current approved immunotherapy combinations, before discussing promising combinatorial approaches in clinical trials and detailing innovative preclinical model systems being used to develop rational combinations. We also discuss the promise of high-order immunotherapy combinations, as well as novel biomarker and combinatorial trial strategies.

Significance:

Although immune-checkpoint inhibitors are approved as dual checkpoint strategies, and in combination with cytotoxic chemotherapy and angiogenesis inhibitors for multiple cancers, patient benefit remains limited. Innovative approaches are required to guide the development of novel immunotherapy combinations, ranging from improvements in preclinical tumor model systems to biomarker-driven trial strategies.

For most of the last century, cancer research was focused on exploiting the antitumor effects of different classes of therapeutics exclusively on cancer cells (1). Over the past decade, however, we have gained a deeper understanding of the role of the immune system in mediating such responses. This has led to a revolution in preclinical, translational, and clinical efforts dedicated to harnessing the immune system for the development of novel immunotherapeutics in cancer medicine (2). Recent studies have led to a vast range of immunotherapies being assessed in clinical trials and subsequently achieving FDA approval.

Immunotherapy has allowed the field of oncology to turn a critical corner, where long-term survival and even durable cures are achievable for patients with metastatic solid tumors. The current reality, however, is that the majority of patients enter care with immune “cold” tumors which respond poorly, if at all, to existing checkpoint therapies (ref. 3; Fig. 1). Immune suppression in these cancers resists reversal with checkpoint blockade due to its multimodal nature encompassing suppressive cytokines, lack of antigen presentation, apoptotic triggering of T cells, and hostile metabolic states and nutrient deprivation. These additional layers of tumor immune privilege must be peeled back therapeutically in order to reveal the benefits of T-cell checkpoint blockade and drive tumor regression. Thus, combinations of multiple immune interventions are necessary to reverse the “cold” tumor state, yet most of the existing “backbone” immunotherapies already approach the ceiling of tolerability even when used at doses that are clearly below their maximum efficacy (4–6).

Figure 1.

Immune-checkpoint blockade in “hot” versus “cold” tumor microenvironments. Immune-checkpoint blockade frees T cells in “hot” tumor microenvironments (top), but fails in “cold” tumors due to dominant, multimodal suppressive mechanisms (bottom).

Figure 1.

Immune-checkpoint blockade in “hot” versus “cold” tumor microenvironments. Immune-checkpoint blockade frees T cells in “hot” tumor microenvironments (top), but fails in “cold” tumors due to dominant, multimodal suppressive mechanisms (bottom).

Close modal

The limitations in efficacy observed with the first wave of approved immunotherapies, primarily involving immune-checkpoint inhibitors, have illustrated a need to improve our understanding of the mechanistic underpinnings of the immune system and thereby develop more robust preclinical and clinical drug development strategies (2). A better understanding of primary and secondary resistance is also required to improve patient outcomes to single-agent immunotherapy strategies (7). Improving our insights into mechanisms of response and resistance is the crucial next step for the future development of immunotherapeutics.

In this review, we begin by detailing the successes observed to date with FDA-approved immunotherapy combinations in different tumor indications, before reviewing promising strategies currently in clinical trial testing. We then describe the use of preclinical models to optimize the development of rational combinations. Finally, we discuss the development of high-order immunotherapy combination strategies and novel biomarker and clinical trial strategies to support the development of combination approaches.

A number of immune checkpoint–based combination treatments are now approved by the FDA (Table 1; Fig. 2), with nivolumab (anti–PD-1) and ipilimumab (anti-CTLA4) the earliest immuno-oncology (IO) combination to receive FDA approval in September 2015 for the first-line treatment of metastatic melanoma (8, 9). Although the treatment of metastatic melanoma had already been transformed by single-agent immune-checkpoint blockade, the nivolumab–ipilimumab combination improved objective response rates (ORR) to 58% and median progression-free survival (mPFS) to 11.5 months compared with nivolumab monotherapy (ORR 45%; mPFS 6.9 months) or ipilimumab alone (ORR 19%; mPFS 2.9 months). Despite a high rate of discontinuation due to toxicities, a survival benefit was apparent even for patients who discontinued treatment, with median overall survival (mOS) not reached at 60 months (10, 11).

Table 1.

FDA-approved immune-checkpoint combinations

Combination Control arm Tumor type(s) No. of patients Grade 3–5 toxicity Median PFS (95% CI; months) Median OS (95% CI; months) 
Metastatic melanoma 
Nivolumab 1 mg/kg and ipilimumab 3 mg/kg (N + I) Nivolumab 1 mg/kg (N)orIpilimumab (3 mg/kg; I) Metastatic melanoma: BRAF wild-type or mutant 945 N + I: 60% N + I: 11.5 (8.7–19.3) N + I: NR (38.2–NR) 
   (n = 314 in combination arm) N: 24% N: 6.9 (5.1–10.2) N: 36.9 (28.2–58.7) 
    I: 28%f I: 2.9 (2.8–3.2) I: 19.9 (16.8–24.6) 
CHECKMATE-067      (8–11) 
Atezolizumab, cobimetinib, vemurafenib (ACV) Cobimetinib, vemurafenib, and placebo (CV) BRAFV600E-mutated advanced melanoma 514 ACV 80% ACV: 15.1 (11.4–18.4) NR 
   (n = 256 in ACV) CV 73%f CV: 10.6 (9.3–12.7) 24 months: 
IMspire150      ACV: 60% 
      CV: 53% (160) 
Colorectal cancer 
Nivolumab 3 mg/kg and ipilimumab 1 mg/kg (N + I) N/A dMMR colorectal cancer, disease progression after 5FU-, irinotecan-, or oxaliplatin-based treatment 119 32% NR NR 
     12 month PFS = 71% (61.4%–78.7%) 12 month OS = 85% (77.0%–90.2%) 
CHECKMATE-142      (13) 
Head and neck squamous cell cancer 
Pembrolizumab and chemotherapy (5-FU and platinum) Cetuximab + chemotherapy (5-FU and platinum; C + chemo) Untreated recurrent or metastatic head and neck squamous cell cancer ITT population: 882 P + chemo: 85% P + chemo: 4.9 (4.7–6.0) C + chemo: 10.7 (9.3–11.7) 
   (n = 281 for P + chemo; n = 301 C + chemo) C + chemo: 83% C + chemo: 5.1 (4.9–6.0) P + chemo: 13.0 (10.9–14.7) 
(P + chemo)KEYNOTE-048   CPS ≥1:  P + chemo: 5.0 (4.7–6.2) C + chemo: 10.4 (9.1–11.7) 
   P + chemo: 242  C + chemo: 5.0 (4.8–5.8) P + chemo: 13.6 (10.7–15.5) 
   C + chemo: 235   (42) 
NSCLC 
Nivolumab 3 mg/kg and ipilimumab 1 mg/kg (N + I) Nivolumab (N), nivolumab + platinum doublet (N + chemo) or chemotherapy (chemo) Stage IV or recurrent NSCLC with WT EGFR and ALK PD-L1% ≥ 1%:1,189 N + I: 36.8% N + I: 5.1 (4.1–6.3) N + I: 17.1 (15.0–20.1) 
    N: 19.9% N: 4.2 (3.0–5.3) N: 15.7 (13.3–18.1) 
   (n = 396 in N; n = 396 in N + I) Chemo: 37.7f Chemo: 5.6 (4.6–5.8) Chemo: 14.9 (12.7–16.7) 
CHECKMATE-227 
   PD-L1% ≤ 1%: 550 N + I: 28.6% N + I: 5.1 (3.2–6.4) N + I: 17.2 (12.8–22.0) 
   (n = 187 for N + I, n = 177 for N + chemo) N + chemo: 58.1% N + chemo: 5.6 (4.6–6.9) N + chemo: 15.2 (12.3–19.8) 
    Chemo: 35.5%f Chemo: 4.7 (4.2–5.6) Chemo: 12.2 (9.2–14.3) (230) 
Nivolumab 3 mg/kg, q3w, ipilimumab 1 mg/kg, q6w + chemo (2 cycles; IO) Chemotherapy: Four cycles (chemo) Stage IV or recurrent NSCLC (EGFR/ALK nonmutated)—treatment naïve IO: 361 IO: 49% NE IO: 15.6 (13.9–20.0) 
   Chemo: 358 Chemo: 40%f  Chemo: 10.9 (9.5–12.5) (231) 
CHECKMATE-9LA 
Pembrolizumab, pemetrexed, and platinum (PPP) Pemetrexed and platinum (chemo) Metastatic nonsquamous NSCLC (EGFR/ALK nonmutated) 616 PPP: 71.9% PPP: 9.0 (8.1–9.9) PPP: 22.0 (19.5–25.2) 
   (n = 410 in IO combination arm) Chemo: 66.8% Chemo: 4.9 (4.7–5.5) Chemo: 10.7 (8.7–13.6) 
KEYNOTE-189      24-month OS =PPP: 45.5% Chemo: 29.9% 
      (22, 24) 
Pembrolizumab, chemotherapy (carboplatin, taxane; PC) Carboplatin and taxane (chemo) Metastatic squamous NSCLC 559 PC: 74.1% PC: 8.0 (6.3–8.4) PC: 17.1 (14.4–19.9) 
   (n = 278 in IO combination arm) Chemo: 69.6% Chemo: 5.1 (4.3–6.0) Chemo: 11.6 (10.1–13.7) 
KEYNOTE-407      (25, 26) 
Chemoradiotherapy—durvalumab consolidation (IO) Chemoradiotherapy—placebo (Pl) Stage III NSCLC 713 IO: 34.9% IO: 17.2 (13.1–23.9) IO: NR (38.4–NR) 
   (n = 476 in IO combination arm) Pl: 32.1%f Pl: 5.6 (4.6–7.7) Pl: 29.1 (22.1–35.1)36-month OS: 
PACIFIC      IO: 57.0% (52.3%–61.4%) 
      Pl: 43.5% (37.0%–49.9%) 
      (27–29) 
Atezolizumab, bevacizumab, carboplatin, and paclitaxel (ABCP)a Bevacizumab, carboplatin, and paclitaxel (BCP) Metastatic non-squamous NSCLC (inc. EGFR/ALK mutated)b Non-EGFR/ALK mutated 1,045 ACP: 43.8% ABCP: 8.3 (7.7–9.8) ABCP: 19.2 (17.0–23.8) 
    ABCP: 58.5% BCP: 6.8 (6.0–7.1) BCP: 14.7 (13.3–16.9) 
   (n = 359 in ABPC, n = 349 in ACP) BCP: 50.0%  (232, 233) 
Or       
Atezolizumab, carboplatin, and paclitaxel (ACP)   EGFR mutated  ABCP: 10.2 (7.9–15.2) ABCP: NE (17.0–NE) 
   124  BCP: 6.9 (5.7–8.5) BCP: 18.7 (11.7–NE) 
IMpower 150   (n = 34 in ABCP and n = 45 in ACP)b  ACP: 6.9 (5.7–8.2) ACP: 21.4 (13.8–NE) 
Atezolizumab, carboplatin, nab-paclitaxel (IO) Platinum-based chemotherapy (chemo) Metastatic non-squamous NSCLC (inc. EGFR/ALK mutated)c Non-EGFR/ALK mutated 723 IO: 74.8% IO: 7.0 (6.2–7.3) IO: 18.6 (16.0–21.2) 
   (n = 451 in IO, n = 228 in chemo) Chemo: 60.8%df Chemo: 5.5 (4.4–5.9) Chemo: 13.9 (12.0–18.7) (234) 
IMpower 130       
Extensive-stage SCLC 
Atezolizumab, carboplatin, and etoposide (APE) Carboplatin and etoposide (chemo) Extensive-stage SCLC 403 APE: 69.2% APE: 5.2 (4.4–5.6) APE: 12.3 (10.8–15.8) 
   (n = 201 in IO combination arm) Chemo: 64.8%f Chemo: 4.3 (4.2–4.5) Chemo: 10.3 (9.3–11.3) (32, 33) 
IMpower 133 
Durvalumab, etoposide, and platinum (DPE) Platinum etoposide (chemo) Extensive-stage SCLC 805 DPE: 66.4%Chemo: DPE: 5.1 (4.7–6.2) DPE: 13.0 (11.5–14.8) 
   (n = 268 for DPE) 68.0%f Chemo: 5.4 (4.8–6.2) Chemo: 10.3 (9.3–11.2) (34) 
CASPIAN 
TNBC 
Atezolizumab and nab-paclitaxel (AP) Nab-paclitaxel (chemo) Advanced TNBC ITT population: 902 AP: 50.1% AP: 7.2 (5.6–7.4) AP: 21.0 (19.0–22.6) 
    Chemo: 43.0%f Chemo: 5.5 (5.3–5.6) Chemo: 17.5 (16.9–20.3) 
IMpassion130   (n = 451 in IO combination arm)    
   PD-L1 ≥ 1%  AP: 7.5 (6.7–9.2) AP: 25.0 (19.6–30.7) 
   369  Chemo: 5.3 (3.8–5.6) Chemo: 18.0 (13.6–20.1)(39, 126) 
   (n = 185 in IO combination arm)    
Renal cell cancer 
Nivolumab 3 mg/kg and ipilimumab 1 mg/kg (N + I) Sunitinib (S) Metastatic clear-cell renal cancer—int.—poor risk (IMDC score) 847 N + I: 48% N + I: 8.2 (8.7–15.5) N + I: NR (35.6–NR) 
   (n = 425 in combination arm) S: 65% S: 8.3 (7.0–8.8) S: 26.6 (22.1–33.4) 
      30-month OS = 
CHECKMATE 214      N + I: 60% (55%–64%) 
      S: 47% (43%–52%) 
  Metastatic clear-cell renal cancer—favorable risk (IMDC score) 249  N + I: 13.9 (9.9–17.9) N + I: NR 
   (n = 125 in combination arm)  S: 19.9 (15.1–23.5) S: 32.9 (NE)30-month OS = 
      N + I: 80% (72%–86%) 
      S: 85% (77%–90%)(16, 17) 
Pembrolizumab and axitinib (PA) Sunitinib (S) Untreated advanced clear-cell renal cell carcinoma 861 PA: 75.8% PA: 15.4 (12.7–18.9) mOS: 
   (n = 432 in IO combination arm) S: 70.6% S: 11.1 (9.1–12.5) PA: NR (NR–NR) 
KEYNOTE-426      S: 35.7 (33.3–NR) 
      24-month OS:PA: 74%S: 66% (51, 235) 
Avelumab and axitinib (A + A) Sunitinib (S) Untreated renal cell carcinoma Overall: 886 A + A: 71.2% Overall population: NE 
   (n = 442 for IO combination) S: 71.5% A + A: 13.3 (11.1–15.3)  
JAVELIN RENAL 101     S: 8.0 (6.7–9.8)  
   PD-L1 ≥ 1%  PD-L1 positive: NE 
   560  A + A: 13.8 (10.1–20.7) (52, 236) 
     S: 7.0 (5.7–9.6)  
Hepatocellular cancer 
Nivolumab 1 mg/kg + ipilimumab 3 mg q4w (A) N/A Sorafenib-treated advanced hepatocellular cancer 148 A: 53%  A: 23 (9–NR) 
   (n = 50 for A, n = 49 for B, n = 49 for C) B: 29%  B: 12 (8–15) 
Nivolumab 1 mg/kg + ipilimumab 3 mg/kg q3w (B) nivolumab 3 mg/kg q2w + ipilimumab 1 mg/kg q6w (C)    C: 31%  C:13 (7–33)(21) 
CHECKMATE 040 
Atezolizumab and bevacizumab (AB) Sorafenib (S) Unresectable hepatocellular cancer 501 AB: 61.1% AB: 6.8 (5.7–8.3) 12-month OS: 
   (n = 336 in IO combination) S: 60.9%f S: 4.3 (4.0–5.6) AB: 67.2% (61.3–73.1) 
      S: 54.6% (45.2–64.0) (53) 
Endometrial cancer 
Pembrolizumab and lenvatinib N/A Advanced endometrial cancer (inc.MSI and MSS)e 108 68.5%f Total: 7.4 (5.3–8.7) Total: 16.7 (15.0–NE) 
     MSI: 18.9 (4.0–NE) MSI: NE (7.4–NE) 
     MSS: 7.4 (5.0–7.6) MSS: 16.4 (13.5–25.9) (44, 45) 
Combination Control arm Tumor type(s) No. of patients Grade 3–5 toxicity Median PFS (95% CI; months) Median OS (95% CI; months) 
Metastatic melanoma 
Nivolumab 1 mg/kg and ipilimumab 3 mg/kg (N + I) Nivolumab 1 mg/kg (N)orIpilimumab (3 mg/kg; I) Metastatic melanoma: BRAF wild-type or mutant 945 N + I: 60% N + I: 11.5 (8.7–19.3) N + I: NR (38.2–NR) 
   (n = 314 in combination arm) N: 24% N: 6.9 (5.1–10.2) N: 36.9 (28.2–58.7) 
    I: 28%f I: 2.9 (2.8–3.2) I: 19.9 (16.8–24.6) 
CHECKMATE-067      (8–11) 
Atezolizumab, cobimetinib, vemurafenib (ACV) Cobimetinib, vemurafenib, and placebo (CV) BRAFV600E-mutated advanced melanoma 514 ACV 80% ACV: 15.1 (11.4–18.4) NR 
   (n = 256 in ACV) CV 73%f CV: 10.6 (9.3–12.7) 24 months: 
IMspire150      ACV: 60% 
      CV: 53% (160) 
Colorectal cancer 
Nivolumab 3 mg/kg and ipilimumab 1 mg/kg (N + I) N/A dMMR colorectal cancer, disease progression after 5FU-, irinotecan-, or oxaliplatin-based treatment 119 32% NR NR 
     12 month PFS = 71% (61.4%–78.7%) 12 month OS = 85% (77.0%–90.2%) 
CHECKMATE-142      (13) 
Head and neck squamous cell cancer 
Pembrolizumab and chemotherapy (5-FU and platinum) Cetuximab + chemotherapy (5-FU and platinum; C + chemo) Untreated recurrent or metastatic head and neck squamous cell cancer ITT population: 882 P + chemo: 85% P + chemo: 4.9 (4.7–6.0) C + chemo: 10.7 (9.3–11.7) 
   (n = 281 for P + chemo; n = 301 C + chemo) C + chemo: 83% C + chemo: 5.1 (4.9–6.0) P + chemo: 13.0 (10.9–14.7) 
(P + chemo)KEYNOTE-048   CPS ≥1:  P + chemo: 5.0 (4.7–6.2) C + chemo: 10.4 (9.1–11.7) 
   P + chemo: 242  C + chemo: 5.0 (4.8–5.8) P + chemo: 13.6 (10.7–15.5) 
   C + chemo: 235   (42) 
NSCLC 
Nivolumab 3 mg/kg and ipilimumab 1 mg/kg (N + I) Nivolumab (N), nivolumab + platinum doublet (N + chemo) or chemotherapy (chemo) Stage IV or recurrent NSCLC with WT EGFR and ALK PD-L1% ≥ 1%:1,189 N + I: 36.8% N + I: 5.1 (4.1–6.3) N + I: 17.1 (15.0–20.1) 
    N: 19.9% N: 4.2 (3.0–5.3) N: 15.7 (13.3–18.1) 
   (n = 396 in N; n = 396 in N + I) Chemo: 37.7f Chemo: 5.6 (4.6–5.8) Chemo: 14.9 (12.7–16.7) 
CHECKMATE-227 
   PD-L1% ≤ 1%: 550 N + I: 28.6% N + I: 5.1 (3.2–6.4) N + I: 17.2 (12.8–22.0) 
   (n = 187 for N + I, n = 177 for N + chemo) N + chemo: 58.1% N + chemo: 5.6 (4.6–6.9) N + chemo: 15.2 (12.3–19.8) 
    Chemo: 35.5%f Chemo: 4.7 (4.2–5.6) Chemo: 12.2 (9.2–14.3) (230) 
Nivolumab 3 mg/kg, q3w, ipilimumab 1 mg/kg, q6w + chemo (2 cycles; IO) Chemotherapy: Four cycles (chemo) Stage IV or recurrent NSCLC (EGFR/ALK nonmutated)—treatment naïve IO: 361 IO: 49% NE IO: 15.6 (13.9–20.0) 
   Chemo: 358 Chemo: 40%f  Chemo: 10.9 (9.5–12.5) (231) 
CHECKMATE-9LA 
Pembrolizumab, pemetrexed, and platinum (PPP) Pemetrexed and platinum (chemo) Metastatic nonsquamous NSCLC (EGFR/ALK nonmutated) 616 PPP: 71.9% PPP: 9.0 (8.1–9.9) PPP: 22.0 (19.5–25.2) 
   (n = 410 in IO combination arm) Chemo: 66.8% Chemo: 4.9 (4.7–5.5) Chemo: 10.7 (8.7–13.6) 
KEYNOTE-189      24-month OS =PPP: 45.5% Chemo: 29.9% 
      (22, 24) 
Pembrolizumab, chemotherapy (carboplatin, taxane; PC) Carboplatin and taxane (chemo) Metastatic squamous NSCLC 559 PC: 74.1% PC: 8.0 (6.3–8.4) PC: 17.1 (14.4–19.9) 
   (n = 278 in IO combination arm) Chemo: 69.6% Chemo: 5.1 (4.3–6.0) Chemo: 11.6 (10.1–13.7) 
KEYNOTE-407      (25, 26) 
Chemoradiotherapy—durvalumab consolidation (IO) Chemoradiotherapy—placebo (Pl) Stage III NSCLC 713 IO: 34.9% IO: 17.2 (13.1–23.9) IO: NR (38.4–NR) 
   (n = 476 in IO combination arm) Pl: 32.1%f Pl: 5.6 (4.6–7.7) Pl: 29.1 (22.1–35.1)36-month OS: 
PACIFIC      IO: 57.0% (52.3%–61.4%) 
      Pl: 43.5% (37.0%–49.9%) 
      (27–29) 
Atezolizumab, bevacizumab, carboplatin, and paclitaxel (ABCP)a Bevacizumab, carboplatin, and paclitaxel (BCP) Metastatic non-squamous NSCLC (inc. EGFR/ALK mutated)b Non-EGFR/ALK mutated 1,045 ACP: 43.8% ABCP: 8.3 (7.7–9.8) ABCP: 19.2 (17.0–23.8) 
    ABCP: 58.5% BCP: 6.8 (6.0–7.1) BCP: 14.7 (13.3–16.9) 
   (n = 359 in ABPC, n = 349 in ACP) BCP: 50.0%  (232, 233) 
Or       
Atezolizumab, carboplatin, and paclitaxel (ACP)   EGFR mutated  ABCP: 10.2 (7.9–15.2) ABCP: NE (17.0–NE) 
   124  BCP: 6.9 (5.7–8.5) BCP: 18.7 (11.7–NE) 
IMpower 150   (n = 34 in ABCP and n = 45 in ACP)b  ACP: 6.9 (5.7–8.2) ACP: 21.4 (13.8–NE) 
Atezolizumab, carboplatin, nab-paclitaxel (IO) Platinum-based chemotherapy (chemo) Metastatic non-squamous NSCLC (inc. EGFR/ALK mutated)c Non-EGFR/ALK mutated 723 IO: 74.8% IO: 7.0 (6.2–7.3) IO: 18.6 (16.0–21.2) 
   (n = 451 in IO, n = 228 in chemo) Chemo: 60.8%df Chemo: 5.5 (4.4–5.9) Chemo: 13.9 (12.0–18.7) (234) 
IMpower 130       
Extensive-stage SCLC 
Atezolizumab, carboplatin, and etoposide (APE) Carboplatin and etoposide (chemo) Extensive-stage SCLC 403 APE: 69.2% APE: 5.2 (4.4–5.6) APE: 12.3 (10.8–15.8) 
   (n = 201 in IO combination arm) Chemo: 64.8%f Chemo: 4.3 (4.2–4.5) Chemo: 10.3 (9.3–11.3) (32, 33) 
IMpower 133 
Durvalumab, etoposide, and platinum (DPE) Platinum etoposide (chemo) Extensive-stage SCLC 805 DPE: 66.4%Chemo: DPE: 5.1 (4.7–6.2) DPE: 13.0 (11.5–14.8) 
   (n = 268 for DPE) 68.0%f Chemo: 5.4 (4.8–6.2) Chemo: 10.3 (9.3–11.2) (34) 
CASPIAN 
TNBC 
Atezolizumab and nab-paclitaxel (AP) Nab-paclitaxel (chemo) Advanced TNBC ITT population: 902 AP: 50.1% AP: 7.2 (5.6–7.4) AP: 21.0 (19.0–22.6) 
    Chemo: 43.0%f Chemo: 5.5 (5.3–5.6) Chemo: 17.5 (16.9–20.3) 
IMpassion130   (n = 451 in IO combination arm)    
   PD-L1 ≥ 1%  AP: 7.5 (6.7–9.2) AP: 25.0 (19.6–30.7) 
   369  Chemo: 5.3 (3.8–5.6) Chemo: 18.0 (13.6–20.1)(39, 126) 
   (n = 185 in IO combination arm)    
Renal cell cancer 
Nivolumab 3 mg/kg and ipilimumab 1 mg/kg (N + I) Sunitinib (S) Metastatic clear-cell renal cancer—int.—poor risk (IMDC score) 847 N + I: 48% N + I: 8.2 (8.7–15.5) N + I: NR (35.6–NR) 
   (n = 425 in combination arm) S: 65% S: 8.3 (7.0–8.8) S: 26.6 (22.1–33.4) 
      30-month OS = 
CHECKMATE 214      N + I: 60% (55%–64%) 
      S: 47% (43%–52%) 
  Metastatic clear-cell renal cancer—favorable risk (IMDC score) 249  N + I: 13.9 (9.9–17.9) N + I: NR 
   (n = 125 in combination arm)  S: 19.9 (15.1–23.5) S: 32.9 (NE)30-month OS = 
      N + I: 80% (72%–86%) 
      S: 85% (77%–90%)(16, 17) 
Pembrolizumab and axitinib (PA) Sunitinib (S) Untreated advanced clear-cell renal cell carcinoma 861 PA: 75.8% PA: 15.4 (12.7–18.9) mOS: 
   (n = 432 in IO combination arm) S: 70.6% S: 11.1 (9.1–12.5) PA: NR (NR–NR) 
KEYNOTE-426      S: 35.7 (33.3–NR) 
      24-month OS:PA: 74%S: 66% (51, 235) 
Avelumab and axitinib (A + A) Sunitinib (S) Untreated renal cell carcinoma Overall: 886 A + A: 71.2% Overall population: NE 
   (n = 442 for IO combination) S: 71.5% A + A: 13.3 (11.1–15.3)  
JAVELIN RENAL 101     S: 8.0 (6.7–9.8)  
   PD-L1 ≥ 1%  PD-L1 positive: NE 
   560  A + A: 13.8 (10.1–20.7) (52, 236) 
     S: 7.0 (5.7–9.6)  
Hepatocellular cancer 
Nivolumab 1 mg/kg + ipilimumab 3 mg q4w (A) N/A Sorafenib-treated advanced hepatocellular cancer 148 A: 53%  A: 23 (9–NR) 
   (n = 50 for A, n = 49 for B, n = 49 for C) B: 29%  B: 12 (8–15) 
Nivolumab 1 mg/kg + ipilimumab 3 mg/kg q3w (B) nivolumab 3 mg/kg q2w + ipilimumab 1 mg/kg q6w (C)    C: 31%  C:13 (7–33)(21) 
CHECKMATE 040 
Atezolizumab and bevacizumab (AB) Sorafenib (S) Unresectable hepatocellular cancer 501 AB: 61.1% AB: 6.8 (5.7–8.3) 12-month OS: 
   (n = 336 in IO combination) S: 60.9%f S: 4.3 (4.0–5.6) AB: 67.2% (61.3–73.1) 
      S: 54.6% (45.2–64.0) (53) 
Endometrial cancer 
Pembrolizumab and lenvatinib N/A Advanced endometrial cancer (inc.MSI and MSS)e 108 68.5%f Total: 7.4 (5.3–8.7) Total: 16.7 (15.0–NE) 
     MSI: 18.9 (4.0–NE) MSI: NE (7.4–NE) 
     MSS: 7.4 (5.0–7.6) MSS: 16.4 (13.5–25.9) (44, 45) 

Abbreviations: IMDC score, International Metastatic Renal Cell Cancer Database Consortium score; int., intermediate; MSI, microsatellite instable; MSS, microsatellite stable; NE, not estimatable; NR, not reached; q2w, every two weeks; q3w, every three weeks; q4w, every four weeks; q6w, every six weeks.

aAnalysis of ACP versus BCP has not yet been reported in NSCLC.

bEGFR- and ALK-mutated nonsquamous NSCLC are excluded from the FDA approval of ABCP.

cEGFR- and ALK-mutated nonsquamous NSCLC are excluded from the FDA approval of atezolizumab, nab-paclitaxel, and carboplatin.

dSafety data include EGFR- and ALK-mutated cancers.

eFDA approval encompasses MSS cancers only.

fGrade 3–4 and grade 5 reported separately in the original paper, percentages calculated by a combination of grade 3–4 and grade 5.

Figure 2.

Different classes of immunotherapy combination strategies. A, Immunotherapy/immunotherapy combinations: example: CTLA4/PD-1 blockade: (1) CTLA4 and PD-1 can no longer suppress T-cell activation, expansion, and effector function; (2) Treg function and differentiation is dampened; (3) phagocytosis of tumor increases from myeloid PD-1 blockade; (4) B7-1/2 can now costimulate T cells through CD28. B, Immunotherapy/chemotherapy combinations: example: gemcitabine/nab-paclitaxel/PD-1 blockade/CD40 agonist: (1) gemcitabine and nab-paclitaxel kill tumor cells releasing tumor antigen; (2) both drugs also selectively deplete myeloid-derived suppressor cells; (3) CD40 activation enhances DC and M1 macrophage activation and increases T-cell priming; (4) activated T cells are protected from attenuation by PD-1 blockade. C, Immunotherapy/adoptive cell therapy combinations: example: anti-CD19 CAR T cells/PD-1 blockade: (1) PD-1 blockade prevents CAR T cells from being rapidly exhausted in the tumor microenvironment; (2) T cell effector function and cytotoxicity are enhanced by PD-1 blockade; (3) PD-1 blockade allows higher levels and duration of IFNγ secretion that maintains an inflamed tumor microenvironment. D, Immunotherapy/targeted therapy combinations: example: VEGFR2/PD-1/CTLA4 blockade: (1) blockade of VEGFR2 normalizes tumor vessels allowing T cells back in; (2) VEGFR2 blockade relieves VEGF inhibition of DC maturation; (3) PD-1 and CTLA4 blockade allows tumor-infiltrating T cells to survive, expand, and kill tumor; (4) T cell–produced IFNγ helps maintain normalized vessels. DC, dendritic cell; TCR, T-cell receptor; TME, tumor microenvironment.

Figure 2.

Different classes of immunotherapy combination strategies. A, Immunotherapy/immunotherapy combinations: example: CTLA4/PD-1 blockade: (1) CTLA4 and PD-1 can no longer suppress T-cell activation, expansion, and effector function; (2) Treg function and differentiation is dampened; (3) phagocytosis of tumor increases from myeloid PD-1 blockade; (4) B7-1/2 can now costimulate T cells through CD28. B, Immunotherapy/chemotherapy combinations: example: gemcitabine/nab-paclitaxel/PD-1 blockade/CD40 agonist: (1) gemcitabine and nab-paclitaxel kill tumor cells releasing tumor antigen; (2) both drugs also selectively deplete myeloid-derived suppressor cells; (3) CD40 activation enhances DC and M1 macrophage activation and increases T-cell priming; (4) activated T cells are protected from attenuation by PD-1 blockade. C, Immunotherapy/adoptive cell therapy combinations: example: anti-CD19 CAR T cells/PD-1 blockade: (1) PD-1 blockade prevents CAR T cells from being rapidly exhausted in the tumor microenvironment; (2) T cell effector function and cytotoxicity are enhanced by PD-1 blockade; (3) PD-1 blockade allows higher levels and duration of IFNγ secretion that maintains an inflamed tumor microenvironment. D, Immunotherapy/targeted therapy combinations: example: VEGFR2/PD-1/CTLA4 blockade: (1) blockade of VEGFR2 normalizes tumor vessels allowing T cells back in; (2) VEGFR2 blockade relieves VEGF inhibition of DC maturation; (3) PD-1 and CTLA4 blockade allows tumor-infiltrating T cells to survive, expand, and kill tumor; (4) T cell–produced IFNγ helps maintain normalized vessels. DC, dendritic cell; TCR, T-cell receptor; TME, tumor microenvironment.

Close modal

Following approval for metastatic melanoma, the combination of nivolumab and ipilimumab has been similarly successful for other tumor sites. Although single-agent immune-checkpoint blockade has been strikingly effective in mismatch repair–deficient (dMMR) cancers (12), this is improved by a combination approach. Nivolumab with lower-dose ipilimumab (1 mg/kg) for the second-line treatment of metastatic dMMR colorectal cancer showed a 60% ORR (13, 14) compared with 31% ORR with nivolumab alone (15). There was a relatively lower toxicity rate than that seen with higher-dose ipilimumab (3 mg/kg) in melanoma, with 32% patients experiencing grade 3 to 5 toxicities.

Similarly, in the first-line treatment of intermediate- or poor-risk advanced clear-cell renal cell cancer, nivolumab–ipilimumab studied in CheckMate 213 resulted in improved patient outcomes, with an ORR of 42%, in contrast to 29% with sunitinib (16, 17). As 1 mg/kg ipilimumab appeared to reduce toxicity without losing efficacy, clinical trials are investigating this dose of ipilimumab with nivolumab (3 mg/kg) in metastatic melanoma, with early data indicating comparable efficacy and lower grade 3 to 5 toxicity rates (18).

Multikinase inhibitors have been the only effective and approved treatments in advanced unresectable hepatocellular cancer (HCC) until the recent approval of immune-checkpoint inhibitors (19). The CheckMate 040 study included comparison of nivolumab–ipilimumab combinations as second-line treatment, comprising arm A (nivolumab 1 mg/kg plus ipilimumab 3 mg/kg every 3 weeks for 4 doses), arm B (nivolumab 3 mg/kg plus ipilimumab 1 mg/kg every 3 weeks for 4 doses followed by maintenance nivolumab every 2 weeks), or arm C (nivolumab 3 mg/kg every 2 weeks plus ipilimumab 1 mg/kg every 6 weeks). ORR was similar between arms A, B, and C at 32%, 31%, and 31%, respectively. Arm A experienced the longest survival at 23 months (95% CI, 9–NA), versus 12 months (95% CI, 9–15), and 13 months (95% CI, 7–33) with arms B and C, respectively. Arms B and C, however, at 1 mg/kg ipilimumab carried the fewest grade 3 to 4 toxicities (29% arm B/31% arm C vs. 53% arm A; ref. 20). FDA approval has been granted to combination IO using the arm A dosing.

In the same trial in HCC, the triplet combination of cabozantinib, nivolumab, and ipilimumab was compared with cabozantinib and nivolumab, including in the first-line setting. Although mOS has not been reported, the triplet arm had an mPFS of 6.8 months versus 5.5 months in the doublet arm, albeit with higher grade 3 to 4 toxicities of 71% versus 42%, respectively (21). Although results from this study are awaited, a number of IO plus other anticancer treatments have already been granted FDA approval, as discussed below.

IO Combinations with Chemotherapy

Combinations with chemotherapy have, to date, been the most numerous approved regimens in various indications. A number of studies in non–small cell lung cancer (NSCLC) have demonstrated a survival advantage when immune-checkpoint blockade is administered in combination with chemotherapy. Pembrolizumab given in combination with pemetrexed and platinum chemotherapy as first-line treatment of nonsquamous NSCLC without EGFR or ALK mutations resulted in an mOS of 22.0 months (95% CI, 19.5–25.2) versus 10.7 months (95% CI 8.7–13.6; refs. 22–24). Similarly, in squamous NSCLC, pembrolizumab in combination with carboplatin and taxane (paclitaxel or nab-paclitaxel) chemotherapy demonstrated an improved mOS at 17.1 months (95% CI, 14.4–19.9) compared with chemotherapy alone (11.6 months; 95% CI, 10.1–13.7; refs. 25, 26). Moreover, following consolidation chemoradiotherapy for stage III NSCLC, durvalumab was shown to lead to a significantly improved mPFS (17.2 vs. 5.6 months) and OS [not reached (NR) vs. 29.1 months], respectively (27–29).

Although combination IO treatments were early successes in the treatment of NSCLC, small cell lung cancer (SCLC) initially proved challenging—studies of maintenance anti–PD-1 and anti-CTLA4–chemotherapy combination treatment did not result in improved PFS (30, 31). However, a study of the PD-L1 inhibitor atezolizumab in combination with carboplatin and etoposide for extensive-stage SCLC resulted in improved mOS of 12.3 months (10.8–15.8) versus 10.3 months (9.3–11.3) in the placebo arm (32, 33), leading to FDA approval in the first-line setting. Similarly, the CASPIAN phase III trial assessed durvalumab in combination with platinum and etoposide chemotherapy as first-line treatment of extensive-stage SCLC, demonstrating an improvement in OS when compared with platinum etoposide alone (13.0 months vs. 10.3 months, HR: 0.73; P = 0.005; ref. 34).

Successful approaches have similarly been reported in breast cancer. Triple-negative breast cancer (TNBC) is the most immunogenic of breast cancers, with high tumor-infiltrating lymphocytes particularly in tumors with features of “BRCAness” (35–37). Initially, however, single-agent anti–PD-1 treatment was disappointing, with a response rate of only 5.3% (38). More recently, atezolizumab in combination with nab-paclitaxel resulted in improved OS (25.0 vs. 18.0 months) in PD-L1–positive tumors (with PD-L1 ≥ 1% on tumor-infiltrating immune cells), although not in unselected TNBC (39). Interestingly, a similar approach with paclitaxel (as opposed to nab-paclitaxel, a regimen that does not require steroid immunosuppression for potential allergic reactions) did not improve patient outcomes (https://www.fda.gov/drugs/resources-information-approved-drugs/fda-issues-alert-about-efficacy-and-potential-safety-concerns-atezolizumab-combination-paclitaxel). These conflicting results highlight the need for robust preclinical studies and the development of rational combinations.

Head and neck squamous cell carcinoma (HNSCC) was an early success in single-agent checkpoint studies, with combination approaches now approved in the first-line HNSCC setting (40). Recurrent or metastatic HNSCC has a dismal prognosis of six to nine months, and standard chemotherapy combination regimens lead to significant comorbidity and involve taxing treatment schedules (41). Pembrolizumab plus chemotherapy was compared with standard-of-care 5-fluorouracil plus platinum chemotherapy with cetuximab given as first-line therapy for incurable or metastatic HNSCC. The immunotherapy combination resulted in improved mOS of 13.0 versus 10.7 months in the standard-of-care group (HR, 0.77; P = 0.0067; ref. 42). This OS benefit was observed despite no significant change in PFS, highlighting the importance of this clinical endpoint in trials of immunotherapy.

IO Combinations with Antiangiogenics

Antiangiogenic agents, targeting vascular endothelial growth factor (VEGF) or its receptor (VEGFR), are promising agents in combination with immune-checkpoint blockade. The increased density of high endothelial venules (typically surrounded by tertiary lymphoid structures) in the tumor microenvironment promotes T-cell trafficking to the tumor and subsequent response to anti–PD-1 therapy (43). This has been borne out in the clinical setting, with many combinations of antiangiogenics and immune-checkpoint blockade now approved. Accelerated FDA approval was granted to the combination of pembrolizumab plus lenvatinib for the treatment of patients with advanced endometrial carcinoma that is not microsatellite instability–high (MSI-H) or dMMR. Efficacy was demonstrated in 108 patients with metastatic endometrial carcinoma that had progressed following at least one prior line of therapy on the KEYNOTE-146 (NCT02501096) trial (44, 45). The overall response rate at 24 weeks was 38.0% [63.6% in patients with MSI-H tumors (n = 11) and 36.2% in patients with microsatellite-stable (MSS) tumors (n = 94)]. For previously treated patients, the overall median duration of response (DOR) was 21.2 months, mPFS was 7.4 months and mOS was 16.7 months. Of note, grade 3 or 4 treatment-related adverse events occurred in 83 of 124 (66.9%) patients.

A quadruple combination regimen [atezolizumab, bevacizumab, carboplatin, and paclitaxel (ABCP)] with the antiangiogenic bevacizumab with atezolizumab (anti–PD-L1) and carboplatin–paclitaxel chemotherapy in nonsquamous NSCLC demonstrated improved mPFS compared with bevacizumab–carboplatin–paclitaxel (8.3 vs. 6.8 months). This study also reported a PFS benefit in patients with EGFR- or ALK-mutant NSCLC, which is recognized to have a lower benefit rate from single-agent PD-1/PD-L1 inhibitors (46–48). The FDA approval, however, did not include the EGFR- and ALK-mutant patient subgroup.

Successful strategies for treating renal cell cancer (RCC) have focused on the sensitivity of RCC to immune-checkpoint inhibitors and antiangiogenic agents, especially given its insensitivity to cytotoxic chemotherapy (49). The combination of bevacizumab with atezolizumab in RCC did not lead to an improvement in OS at interim analysis (33.6 months vs 34.9 months) despite demonstrating a superior mPFS versus sunitinib alone (11.2 months vs. 8.4 months, P < 0.02; ref. 50). However, single-agent checkpoint inhibition has also been added to the oral small-molecule axitinib in two phase III trials. Compared with sunitinib, the combination of pembrolizumab and axitinib led to a benefit in 12-month OS of 89.9% versus 78.3% (HR, 0.53; 95% CI, 0.38–0.74; P < 0.01; ref. 51). Axitinib has also been assessed in combination with avelumab in untreated RCC and compared with sunitinib. Although OS for this combination is yet to be reported, there was a PFS benefit of 13.8 months (95% CI, 11.1–NR) versus 8.4 months (6.9–11.1) with sunitinib (52). Both combinations have subsequently been approved by the FDA.

In the IMBrave150 open-label phase III randomized trial in patients with resectable HCC naïve to systemic therapy, the combination of the angiogenesis inhibitor bevacizumab added to atezolizumab was compared with sorafenib in a 2:1 fashion. Although the mOS has not been published, the OS at 12 months was 67.2% for the IO combination versus 54.6% for sorafenib. mPFS was improved in the IO group [6.8 months (95% CI, 5.7–8.3) versus 4.3 months (95% CI, 4.0–5.6)], respectively. Grade 3 or 4 adverse events were comparable between both arms, at 56.5% with the IO combination and 55.1% with sorafenib (53).

Also in HCC, the phase Ib/II trial combining lenvatinib and pembrolizumab regardless of PD-L1 status resulted in an ORR of 36.4% (8/22) but with 91% experiencing grade 3 or 4 toxicities (54). This combination is now undergoing evaluation in a randomized phase III study (NCT04199104).

IO Combinations with Targeted Agents

The combination of BRAF and MEK inhibitors for the treatment of BRAF-mutant metastatic melanoma transformed outcomes in this aggressive subgroup, achieving rapid disease control, and is now also approved in the adjuvant setting (55–57). In IMspire150, vemurafenib and cobimetinib given in combination with atezolizumab improved PFS for patients with unresectable or metastatic melanoma (15.1 months; 95% CI, 11.4–18.4) versus vemurafenib and cobimetinib with placebo (10.6 months; 95% CI, 9.3–12.7). DOR was also improved with the addition of checkpoint inhibition [21.0 (95% CI, 15.1–NE) vs. 12.6 months (95% CI, 10.5–16.6)] with, importantly, little change in the grade 3 to 4 toxicity rate (79% vs. 73%). FDA approval was granted in July 2020, with a first cycle of vemurafenib and cobimetinib, and atezolizumab from cycle 2 onward. Interestingly, a subsequent phase III trial of the anti–PD-1 antibody spartalizumab in combination with the BRAF inhibitor dabrafenib and MEK inhibitor trametinib in patients with BRAF-mutated metastatic melanoma, reported at the 2020 European Society for Medical Oncology (ESMO) Annual Meeting, failed to reach its primary endpoint and raised questions regarding the utility of triplet combinations in melanoma (58).

These encouraging results with regulatory approval in multiple indications with combination immunotherapy strategies have provided the continued impetus for a large number of combination studies currently in preclinical trial testing, and selected studies are detailed in Table 2.

Table 2.

Selected preclinical studies of IO combinations

ICBCombinationModelTreatment scheduleKey resultsReferences
PD-1 Targeting ANGPT2 and VEGF Breast cancer (MMTV-PyMT and 4T1), colon cancer (MC38-OVA), and melanoma (B16-OVA) Anti–PD-1 (RMP1-14; 10 mg/kg), anti-ANGPT2 (LC06, 10 mg/kg), and anti-VEGF (B20-4.1, 10 mg/kg), once per week; concurrent treatment Antiangiogenic treatment induced PD-L1 expression in tumor endothelial cells.The combination enhanced activation and cytokine production of CD8+ T cells. 237 
PD-1 Targeting VEGF Colon cancer (CT26) Anti–PD-1 (RMP1-14; 0.25 mg/kg twice a week), and anti-VEGF (Genentech); concurrent treatment VEGF enhanced the expression of inhibitory immune checkpoints (PD-1, CTLA4, TIM3, and LAG3) on CD8+ T cells.VEGF mediated resistance to anti–PD-1 treatment.Anti-VEGF synergized with anti–PD-1 treatment in controlling tumor growth. 238 
PD-1 Targeting ANG2 and VEGF Breast cancer (EMT-6/CDDP) Anti–PD-L1 (10F.9G2; 5 mg/kg; twice a week), and anti-VEGF/Ang (CVX-241; 30 mg/kg/week); concurrent treatment The combination slightly extended mice survival. 239 
PD-1/PD-L1 Cytotoxic chemotherapy Bladder cancer (MBT-2 and MB49), breast cancer (4T1), and colon cancer (MC38) Anti–PD-1/anti–PD-L1 (RMP1-14/10F.9G2; 12.5 mg/kg; once per week); cytotoxic reagents including methotrexate + vinblastine + doxorubicin + cisplatin (MB49 and MBT-2), cyclophosphamide + doxorubicin (4T1), capecitabine + oxaliplatin (MC38); concurrent treatment The combination of cytotoxic chemotherapy with ICIs may result in enhanced (MC38 and MB49), similar (4T1), or reduced antitumor activity (MBT-2), in a model- and regimen-dependent fashion. 240 
PD-1/CTLA4 Cytotoxic chemotherapy Mesothelioma (RN5) Anti–PD-1/anti-CTLA4 (RMP1-14/UC10-4F10-11; 250 μg; once per week), gemcitabine; concurrent treatment The combination significantly extended the survival of tumor-bearing mice and increased tumor infiltration of immune cells. 241 
ICOS Cytotoxic chemotherapy Breast cancer (MDA-MB-231) in humanized NSG mice Anti-ICOS (314.8; 250 μg; once per week), cyclophosphamide; concurrent treatment This combination reduced tumor growth and increased the ratio of CD8+/Treg. Depletion of human CD8+ T cells or of murine myeloid cells blunted antitumor effect of this combination treatment. 242 
CTLA4 Targeting DNA methyltransferase Ovarian cancer (BR5FVB1-Akt) Anti-CTLA4 (9H10), decitabine; concurrent treatment This combination increased the percentage of effector memory T cells at tumor sites, enhanced the cytokine production by T cells and NK cells, and prolonged the duration of high cytokine production by immune cells. 243 
PD-1 Targeting DNA methyl-transferase and histone deacetylase Ovarian cancer (ID8) Anti–PD-1 (200 μg twice per week for two weeks), Vidaza (AZA), entinostat (MS17), and/or givinostat (ITF17); pretreatment of epigenetic reagents for two weeks followed by the combination of anti–PD-1 MS17/ITF17 + α-PD-1 is ineffective, whereas AZA + α-PD-1 is effective.The longest OS was found in the group treated with AZA, ITF17, and anti–PD-1.Through upregulating type I IFN signaling, AZA + ITF17 treatment increased T- and NK-cell activation, and reduced the number of macrophages. 244 
PD-L1 Targeting EZH2 and DNMT1 Ovarian cancer (ID8) Anti–PD-L1 (10F.9G2, 10 mg/kg twice per week for two weeks); concurrent treatment The triple combination displayed synergistic antitumor effect and increased the number of tumor-infiltrating CD8+ T cells and Th1-type chemokine expression. 245 
CTLA4 Oncolytic vaccinia virus Colon cancer (MC38), kidney cancer (Renca) Anti-CTLA4 (9D9, 100 μg, twice per week for two weeks), double-deleted Western reserve viral strains; concurrent and sequential Delayed anti-CTLA4 treatment enhanced the antitumor effects of the oncolytic vaccinia virus, which are dependent on CD8+ and NK cells, but not CD4+ cells. Concurrent treatment did not improve the efficacy of oncolytic vaccinia. 246 
PD-1/CTLA4 Oncolytic herpes simplex virus (oHSV) Glioma cells (005 GSCs and CT-2A) Anti–PD-1 and anti-CTLA4 (RMP1-14/9D9, 10 and 5 mg/kg respectively, twice per week for two weeks), oHSV-mIL12; concurrent The triple combination significantly extended survival, facilitated M1-like polarization of macrophages, and increased the ratio of Teff/Treg.CD4+ and CD8+ T cells as well as macrophages are required for synergistic activity. 247 
PD-1/CTLA4 Maraba oncolytic virus (oHSV) Breast cancer (4T1, EM6, and E0771) Anti–PD-1 and anti-CTLA4 (RMP1-14/9D9, 100 μg, every two days for a total of five injections), Maraba treatment first, then antibody treatment Maraba treatment sensitized breast tumor cells to immune-checkpoint blockade. 248 
PD-1 Aglatimagene besadenovec (Adv-tk) Glioblastoma (GL26 and CT-2) α-PD-1 (29F.1A12; 200 μg; every two days for a total of four injections), intratumoral injection of Adv-tk; concurrent treatment Anti–PD-1 enhanced the effectiveness of this oncolytic viral treatment by overcoming IFN-induced PD-L1–mediated inhibitory signals. 249 
PD-1/CTLA4 Oncolytic vacciniavirus Kidney cancer (renca) and breast cancer (MMTV-PyMT transgenic mice) Anti–PD-1 and anti-CTLA4 (J43/9D9, 10 and 4 mg/kg respectively, every three days), mJX-594 (JX), a targeted and GM-CSF–armed oncolytic vaccinia virus; concurrent versus sequential treatment This combination delayed tumor growth and increased the number and effector function of tumor-infiltrating T cells regardless of the treatment schedule. 250 
PD-1/CD137/CD40 Stereotactic body radiation (SBRT) Breast cancer (4T1.2 and AT-3) Anti-CD137 (100 μg) and/or anti-CD40 (100 μg), anti–PD-1 (100 μg) on days 0, 4, 8, and 12 relative to radiotherapy, 12 Gy radiotherapy Tumor eradication can be achieved by the triple combination of IR, anti–PD-1, and anti-CD137, but not the combination of IR, anti-CD40, and anti-CD137.The profound therapeutic efficacy of the triple combination of IR + anti–PD-1 + anti-CD137 is associated with the capacity of radiotherapy to enrich the tumor microenvironment for critical tumor-reactive CD8+ T cells that coexpress CD137 and PD-1. 251 
PD-L1/CTLA4 SBRT Melanoma (B16), pancreatic cancer (PDA.4662) and breast cancer (TSA) Anti–PD-L1 and anti-CTLA4 (10F.9G2/9H10, 200 μg, two days before tumor inoculation and every four days); concurrent or sequential treatment Sequential treatment of anti-CTLA4 and SBRT has similar antitumor effect as concurrent treatment.Adding anti–PD-L1 reinvigorated tumor-infiltrating T cells and synergized with the combination of anti-CTLA4 and SBRT. 252 
PD-1/4-1BB SBRT Braf-mutant and PTEN-deficient melanoma GEMM Anti–PD-1 and anti-4-1BB (RMP1-14/3H3, 100 μg, twice per week for two weeks); SBRT first followed by antibody treatment The triple combination significantly prolonged OS by increasing immunogenicity of tumors. 253 
PD-1/CTLA4 Irreversible electroporation (IRE) Pancreatic cancer (KRAS*) Anti–PD-1 and anti-CTLA4 (J43/9D9, 100 μg, every two days for a total of six injections); IRE first followed by antibody treatment The combination of IRE and anti–PD-1 promoted tumor infiltration by CD8+ cells and enhanced long-term memory immune response.Adding anti-CTLA4 to this double combination cannot further improve antitumor activity. 254 
PD-1 Adoptive T-cell therapy Melanoma (B16) and colon cancer (MC38) Anti–PD-1 (RMP1-14; 100 μg; twice a week), IL2 and gp100-pulsed DCs; concurrent treatment Anti–PD-1 treatment enhances tumor trafficking of transferred T cells via elevating IFNγ-inducible chemokines. 255 
PD-1 Adoptive T-cell therapy Human lung cancer (A549) Anti–PD-1 (10 mg/Kg; every five days), human T cells engineered to target NY-ESO-1; concurrent treatment Anti–PD-1 treatment augmented antitumor activity of transferred tumor-reactive T cells by enhancing effector function of T cells. 256 
PD-1/CTLA4 Targeting hypoxia Prostate cancer (TRAMP-c2 and a PTEN and SMAD4-deficient GEMM) Anti–PD-1 (RMP1-14, 250 μg) and anti-CTLA4 (9H10, 100 μg, every three days for a total of six injections), TH-302 (evofosfamide); concurrent treatment The triple combination can achieve profound antitumor activity by enhancing the ratio of T cells/MDSCs. 257 
PD-L1 Targeting CDK4/6 Breast cancer (an MMTV-rtTA/tetO-HER2 GEMM) α-PD-L1 (10F.9G2, 200 μg, every three days), abemaciclib/palbociclib; concurrent treatment CDK4/6 inhibition synergizes with anti–PD-1 by stimulating the IFN signaling pathway. 258 
PD-1/CTLA4 Targeting KIT Colon cancer (Colon 26) Anti–PD-1 (5 mg/kg on days 3, 6, 10, and 13), anti-CTLA4 (5 mg/kg on day 8, 2.5 mg/kg on days 11 and 14), anti-KIT (1, 3, 10, or 15 mg/kg on days 3, 6, 10, and 13) The combination of anti-KIT and anti-CTLA4 reduced tumor growth by reducing the number of M-MDSCs.The combination of anti-KIT with anti–PD-1 delayed tumor development, but without statistical significance. 259 
PD-1/PD-L1 Targeting KIT Gastrointestinal stromal cancer (a KitV558Δ/+ GEMM) Anti–PD-1 (RMP1-14, 250 μg) or anti-PD-L1 (10F.9G2, 200 μg) every four days for a total of four treatments, imatinib; concurrent treatment Combining imatinib with either anti–PD-1 or anti–PD-L1 can achieve better antitumor activity by suppressing multiple immunosuppressive mechanisms, including IDO pathway. 260 
PD-1/CTLA4 Targeting PI3Kβ A Braf-mutant and PTEN-deficient melanoma GEMM Anti–PD-1 (29F.1A12, 100 μg)/anti-CTLA4 (9H10, 100 μg) every two days for a total of three treatments, GSK2636771; concurrent treatment GSK2636771 synergizes with ICIs by enhancing tumor infiltration of T cells and reducing VEGF expression. 187 
PD-1/CTLA4 Targeting PI3Kγ Melanoma and breast cancer (B16-GM-CSF and 4T1) Anti–PD-1 (RMP1–14, 250 μg)/anti-CTLA4 (9H10, 100 μg) every three days for a total of six treatments, IPI-549; concurrent treatment The triple combination significantly improved OS of tumor-bearing mice. 261 
PD-1 Targeting PI3Kγ Head and neck squamous cell cancer (MEER and SCCVII) α-PD-1 (RMP1-14, 250 μg) every four days for a total of four treatments, IPI-549; concurrent treatment This combination inhibited the expression of immunosuppressive genes, promoted CD8+ T-cell recruitment, and induced sustained tumor regression. 262 
PD-1/CD137/CD134 Targeting BRAF and MEK Melanoma (SM1) α-PD-1/α-PD-L1/α-CD137/α-CD134 (200 μg) every four days for a total of four treatments, dabrafenib and trametinib; concurrent Adding immune-stimulating reagents such as α-CD137 and α-CD134 can further improve the efficacy of the combination of dabrafenib, trametinib, and PD-1 inhibition. 263 
PD-1 Targeting PI3K Breast cancer (MMTV-PyMT and 4T1) α-PD-1 (100 μg) twice per week; BKM120 and genetic approach; concurrent treatment Inhibiting the PI3K pathway improved tumor sensitivity of α-PD-1 treatment and increased tumor infiltration of T cells. 264 
PD-1 Targeting COX2 pathway Melanoma (a cell line derived from a Braf+/LSLV600E; Tyr::CreERT2+/o;p16INK4a/mouse) α-PD-1 (RMP1-14, 200 μg)/twice per week for two weeks, a genetic approach to silencing PGES1; concurrent treatment Inhibiting PGES1, an effector of the COX2 pathway, enhanced the sensitivity of tumor α-PD-1 treatment. 265 
PD-1/CTLA4 Targeting MEK Kras-mutant NSCLC (LLC) α-PD-1(RMP1-14, 250 μg)/α-CTLA4 (9H10 or 9D9, 100 μg) twice per week for three weeks, selumetinib (continuous treatment versus pulsatile treatment); concurrent treatment Only the combination of CTLA4 blockade with pulsatile MEKi can significantly prolong OS of tumor-bearing mice. This synergistic effect is dependent on immune cells. 266 
PD-1 Targeting PARP BRCA1-deficient ovarian cancer (a Trp53−/−, Brca1−/−,Myc GEMM) α-PD-1(332.8H3, 250 μg) every three days, olaparib; concurrent treatment PARP inhibition augmented antitumor activity of α-PD-1 through a STING-dependent manner. 142 
PD-1/CD134 STING-based neoantigen vaccine Pancreatic cancer (Pan02) Anti–PD-1 (RMP1-14, 100 μg), α-OX40 (OX86, 50 μg) ADU-V16 (sting adjuvant), peptide vaccine; concurrent treatment The triple combination can induce a durable antitumor immune response and eliminate majority of tumor development. 267 
CTLA4 Neoantigen vaccine Pancreatic cancer (Pan02) Anti–CTLA4 (9H10, 100 μg, every two days for a total of three treatments), pIC, and anti-CD40 (adjuvant), peptide vaccine; concurrent treatment Anti–CTLA4 significantly increased the number of vaccine-induced effector T cells within the tumor, and this effect is dependent on T cell–derived IL3. 268 
ICBCombinationModelTreatment scheduleKey resultsReferences
PD-1 Targeting ANGPT2 and VEGF Breast cancer (MMTV-PyMT and 4T1), colon cancer (MC38-OVA), and melanoma (B16-OVA) Anti–PD-1 (RMP1-14; 10 mg/kg), anti-ANGPT2 (LC06, 10 mg/kg), and anti-VEGF (B20-4.1, 10 mg/kg), once per week; concurrent treatment Antiangiogenic treatment induced PD-L1 expression in tumor endothelial cells.The combination enhanced activation and cytokine production of CD8+ T cells. 237 
PD-1 Targeting VEGF Colon cancer (CT26) Anti–PD-1 (RMP1-14; 0.25 mg/kg twice a week), and anti-VEGF (Genentech); concurrent treatment VEGF enhanced the expression of inhibitory immune checkpoints (PD-1, CTLA4, TIM3, and LAG3) on CD8+ T cells.VEGF mediated resistance to anti–PD-1 treatment.Anti-VEGF synergized with anti–PD-1 treatment in controlling tumor growth. 238 
PD-1 Targeting ANG2 and VEGF Breast cancer (EMT-6/CDDP) Anti–PD-L1 (10F.9G2; 5 mg/kg; twice a week), and anti-VEGF/Ang (CVX-241; 30 mg/kg/week); concurrent treatment The combination slightly extended mice survival. 239 
PD-1/PD-L1 Cytotoxic chemotherapy Bladder cancer (MBT-2 and MB49), breast cancer (4T1), and colon cancer (MC38) Anti–PD-1/anti–PD-L1 (RMP1-14/10F.9G2; 12.5 mg/kg; once per week); cytotoxic reagents including methotrexate + vinblastine + doxorubicin + cisplatin (MB49 and MBT-2), cyclophosphamide + doxorubicin (4T1), capecitabine + oxaliplatin (MC38); concurrent treatment The combination of cytotoxic chemotherapy with ICIs may result in enhanced (MC38 and MB49), similar (4T1), or reduced antitumor activity (MBT-2), in a model- and regimen-dependent fashion. 240 
PD-1/CTLA4 Cytotoxic chemotherapy Mesothelioma (RN5) Anti–PD-1/anti-CTLA4 (RMP1-14/UC10-4F10-11; 250 μg; once per week), gemcitabine; concurrent treatment The combination significantly extended the survival of tumor-bearing mice and increased tumor infiltration of immune cells. 241 
ICOS Cytotoxic chemotherapy Breast cancer (MDA-MB-231) in humanized NSG mice Anti-ICOS (314.8; 250 μg; once per week), cyclophosphamide; concurrent treatment This combination reduced tumor growth and increased the ratio of CD8+/Treg. Depletion of human CD8+ T cells or of murine myeloid cells blunted antitumor effect of this combination treatment. 242 
CTLA4 Targeting DNA methyltransferase Ovarian cancer (BR5FVB1-Akt) Anti-CTLA4 (9H10), decitabine; concurrent treatment This combination increased the percentage of effector memory T cells at tumor sites, enhanced the cytokine production by T cells and NK cells, and prolonged the duration of high cytokine production by immune cells. 243 
PD-1 Targeting DNA methyl-transferase and histone deacetylase Ovarian cancer (ID8) Anti–PD-1 (200 μg twice per week for two weeks), Vidaza (AZA), entinostat (MS17), and/or givinostat (ITF17); pretreatment of epigenetic reagents for two weeks followed by the combination of anti–PD-1 MS17/ITF17 + α-PD-1 is ineffective, whereas AZA + α-PD-1 is effective.The longest OS was found in the group treated with AZA, ITF17, and anti–PD-1.Through upregulating type I IFN signaling, AZA + ITF17 treatment increased T- and NK-cell activation, and reduced the number of macrophages. 244 
PD-L1 Targeting EZH2 and DNMT1 Ovarian cancer (ID8) Anti–PD-L1 (10F.9G2, 10 mg/kg twice per week for two weeks); concurrent treatment The triple combination displayed synergistic antitumor effect and increased the number of tumor-infiltrating CD8+ T cells and Th1-type chemokine expression. 245 
CTLA4 Oncolytic vaccinia virus Colon cancer (MC38), kidney cancer (Renca) Anti-CTLA4 (9D9, 100 μg, twice per week for two weeks), double-deleted Western reserve viral strains; concurrent and sequential Delayed anti-CTLA4 treatment enhanced the antitumor effects of the oncolytic vaccinia virus, which are dependent on CD8+ and NK cells, but not CD4+ cells. Concurrent treatment did not improve the efficacy of oncolytic vaccinia. 246 
PD-1/CTLA4 Oncolytic herpes simplex virus (oHSV) Glioma cells (005 GSCs and CT-2A) Anti–PD-1 and anti-CTLA4 (RMP1-14/9D9, 10 and 5 mg/kg respectively, twice per week for two weeks), oHSV-mIL12; concurrent The triple combination significantly extended survival, facilitated M1-like polarization of macrophages, and increased the ratio of Teff/Treg.CD4+ and CD8+ T cells as well as macrophages are required for synergistic activity. 247 
PD-1/CTLA4 Maraba oncolytic virus (oHSV) Breast cancer (4T1, EM6, and E0771) Anti–PD-1 and anti-CTLA4 (RMP1-14/9D9, 100 μg, every two days for a total of five injections), Maraba treatment first, then antibody treatment Maraba treatment sensitized breast tumor cells to immune-checkpoint blockade. 248 
PD-1 Aglatimagene besadenovec (Adv-tk) Glioblastoma (GL26 and CT-2) α-PD-1 (29F.1A12; 200 μg; every two days for a total of four injections), intratumoral injection of Adv-tk; concurrent treatment Anti–PD-1 enhanced the effectiveness of this oncolytic viral treatment by overcoming IFN-induced PD-L1–mediated inhibitory signals. 249 
PD-1/CTLA4 Oncolytic vacciniavirus Kidney cancer (renca) and breast cancer (MMTV-PyMT transgenic mice) Anti–PD-1 and anti-CTLA4 (J43/9D9, 10 and 4 mg/kg respectively, every three days), mJX-594 (JX), a targeted and GM-CSF–armed oncolytic vaccinia virus; concurrent versus sequential treatment This combination delayed tumor growth and increased the number and effector function of tumor-infiltrating T cells regardless of the treatment schedule. 250 
PD-1/CD137/CD40 Stereotactic body radiation (SBRT) Breast cancer (4T1.2 and AT-3) Anti-CD137 (100 μg) and/or anti-CD40 (100 μg), anti–PD-1 (100 μg) on days 0, 4, 8, and 12 relative to radiotherapy, 12 Gy radiotherapy Tumor eradication can be achieved by the triple combination of IR, anti–PD-1, and anti-CD137, but not the combination of IR, anti-CD40, and anti-CD137.The profound therapeutic efficacy of the triple combination of IR + anti–PD-1 + anti-CD137 is associated with the capacity of radiotherapy to enrich the tumor microenvironment for critical tumor-reactive CD8+ T cells that coexpress CD137 and PD-1. 251 
PD-L1/CTLA4 SBRT Melanoma (B16), pancreatic cancer (PDA.4662) and breast cancer (TSA) Anti–PD-L1 and anti-CTLA4 (10F.9G2/9H10, 200 μg, two days before tumor inoculation and every four days); concurrent or sequential treatment Sequential treatment of anti-CTLA4 and SBRT has similar antitumor effect as concurrent treatment.Adding anti–PD-L1 reinvigorated tumor-infiltrating T cells and synergized with the combination of anti-CTLA4 and SBRT. 252 
PD-1/4-1BB SBRT Braf-mutant and PTEN-deficient melanoma GEMM Anti–PD-1 and anti-4-1BB (RMP1-14/3H3, 100 μg, twice per week for two weeks); SBRT first followed by antibody treatment The triple combination significantly prolonged OS by increasing immunogenicity of tumors. 253 
PD-1/CTLA4 Irreversible electroporation (IRE) Pancreatic cancer (KRAS*) Anti–PD-1 and anti-CTLA4 (J43/9D9, 100 μg, every two days for a total of six injections); IRE first followed by antibody treatment The combination of IRE and anti–PD-1 promoted tumor infiltration by CD8+ cells and enhanced long-term memory immune response.Adding anti-CTLA4 to this double combination cannot further improve antitumor activity. 254 
PD-1 Adoptive T-cell therapy Melanoma (B16) and colon cancer (MC38) Anti–PD-1 (RMP1-14; 100 μg; twice a week), IL2 and gp100-pulsed DCs; concurrent treatment Anti–PD-1 treatment enhances tumor trafficking of transferred T cells via elevating IFNγ-inducible chemokines. 255 
PD-1 Adoptive T-cell therapy Human lung cancer (A549) Anti–PD-1 (10 mg/Kg; every five days), human T cells engineered to target NY-ESO-1; concurrent treatment Anti–PD-1 treatment augmented antitumor activity of transferred tumor-reactive T cells by enhancing effector function of T cells. 256 
PD-1/CTLA4 Targeting hypoxia Prostate cancer (TRAMP-c2 and a PTEN and SMAD4-deficient GEMM) Anti–PD-1 (RMP1-14, 250 μg) and anti-CTLA4 (9H10, 100 μg, every three days for a total of six injections), TH-302 (evofosfamide); concurrent treatment The triple combination can achieve profound antitumor activity by enhancing the ratio of T cells/MDSCs. 257 
PD-L1 Targeting CDK4/6 Breast cancer (an MMTV-rtTA/tetO-HER2 GEMM) α-PD-L1 (10F.9G2, 200 μg, every three days), abemaciclib/palbociclib; concurrent treatment CDK4/6 inhibition synergizes with anti–PD-1 by stimulating the IFN signaling pathway. 258 
PD-1/CTLA4 Targeting KIT Colon cancer (Colon 26) Anti–PD-1 (5 mg/kg on days 3, 6, 10, and 13), anti-CTLA4 (5 mg/kg on day 8, 2.5 mg/kg on days 11 and 14), anti-KIT (1, 3, 10, or 15 mg/kg on days 3, 6, 10, and 13) The combination of anti-KIT and anti-CTLA4 reduced tumor growth by reducing the number of M-MDSCs.The combination of anti-KIT with anti–PD-1 delayed tumor development, but without statistical significance. 259 
PD-1/PD-L1 Targeting KIT Gastrointestinal stromal cancer (a KitV558Δ/+ GEMM) Anti–PD-1 (RMP1-14, 250 μg) or anti-PD-L1 (10F.9G2, 200 μg) every four days for a total of four treatments, imatinib; concurrent treatment Combining imatinib with either anti–PD-1 or anti–PD-L1 can achieve better antitumor activity by suppressing multiple immunosuppressive mechanisms, including IDO pathway. 260 
PD-1/CTLA4 Targeting PI3Kβ A Braf-mutant and PTEN-deficient melanoma GEMM Anti–PD-1 (29F.1A12, 100 μg)/anti-CTLA4 (9H10, 100 μg) every two days for a total of three treatments, GSK2636771; concurrent treatment GSK2636771 synergizes with ICIs by enhancing tumor infiltration of T cells and reducing VEGF expression. 187 
PD-1/CTLA4 Targeting PI3Kγ Melanoma and breast cancer (B16-GM-CSF and 4T1) Anti–PD-1 (RMP1–14, 250 μg)/anti-CTLA4 (9H10, 100 μg) every three days for a total of six treatments, IPI-549; concurrent treatment The triple combination significantly improved OS of tumor-bearing mice. 261 
PD-1 Targeting PI3Kγ Head and neck squamous cell cancer (MEER and SCCVII) α-PD-1 (RMP1-14, 250 μg) every four days for a total of four treatments, IPI-549; concurrent treatment This combination inhibited the expression of immunosuppressive genes, promoted CD8+ T-cell recruitment, and induced sustained tumor regression. 262 
PD-1/CD137/CD134 Targeting BRAF and MEK Melanoma (SM1) α-PD-1/α-PD-L1/α-CD137/α-CD134 (200 μg) every four days for a total of four treatments, dabrafenib and trametinib; concurrent Adding immune-stimulating reagents such as α-CD137 and α-CD134 can further improve the efficacy of the combination of dabrafenib, trametinib, and PD-1 inhibition. 263 
PD-1 Targeting PI3K Breast cancer (MMTV-PyMT and 4T1) α-PD-1 (100 μg) twice per week; BKM120 and genetic approach; concurrent treatment Inhibiting the PI3K pathway improved tumor sensitivity of α-PD-1 treatment and increased tumor infiltration of T cells. 264 
PD-1 Targeting COX2 pathway Melanoma (a cell line derived from a Braf+/LSLV600E; Tyr::CreERT2+/o;p16INK4a/mouse) α-PD-1 (RMP1-14, 200 μg)/twice per week for two weeks, a genetic approach to silencing PGES1; concurrent treatment Inhibiting PGES1, an effector of the COX2 pathway, enhanced the sensitivity of tumor α-PD-1 treatment. 265 
PD-1/CTLA4 Targeting MEK Kras-mutant NSCLC (LLC) α-PD-1(RMP1-14, 250 μg)/α-CTLA4 (9H10 or 9D9, 100 μg) twice per week for three weeks, selumetinib (continuous treatment versus pulsatile treatment); concurrent treatment Only the combination of CTLA4 blockade with pulsatile MEKi can significantly prolong OS of tumor-bearing mice. This synergistic effect is dependent on immune cells. 266 
PD-1 Targeting PARP BRCA1-deficient ovarian cancer (a Trp53−/−, Brca1−/−,Myc GEMM) α-PD-1(332.8H3, 250 μg) every three days, olaparib; concurrent treatment PARP inhibition augmented antitumor activity of α-PD-1 through a STING-dependent manner. 142 
PD-1/CD134 STING-based neoantigen vaccine Pancreatic cancer (Pan02) Anti–PD-1 (RMP1-14, 100 μg), α-OX40 (OX86, 50 μg) ADU-V16 (sting adjuvant), peptide vaccine; concurrent treatment The triple combination can induce a durable antitumor immune response and eliminate majority of tumor development. 267 
CTLA4 Neoantigen vaccine Pancreatic cancer (Pan02) Anti–CTLA4 (9H10, 100 μg, every two days for a total of three treatments), pIC, and anti-CD40 (adjuvant), peptide vaccine; concurrent treatment Anti–CTLA4 significantly increased the number of vaccine-induced effector T cells within the tumor, and this effect is dependent on T cell–derived IL3. 268 

Abbreviations: ICI, immune checkpoint inhibitor; pIC, polyinosinic:polycytidylic acid.

The generally safe and durable responses observed with single-agent PD-1/PD-L1 inhibitors have established this class of agents as the therapeutic backbone of the majority of immune-checkpoint inhibitor combinations (59). The tantalizing promise of durable antitumor responses has understandably fed enthusiasm for further combination studies, with 2,251 active PD-1/PD-L1 inhibitor–based combination trials of 295 targets in 2019 (www.cancerresearch.org; Fig. 2).

In metastatic melanoma, high-dose anti-CTLA4 agent ipilimumab at 10 versus 3 mg/kg was found to have increased mOS (15.7 months vs. 11.5 months; P = 0.04) at a significant cost of increased treatment-related adverse events (37% vs. 18%; ref. 5). Conversely, the anti–PD-1 pembrolizumab has been found to have similar efficacy with similar rates of toxicity at two different weight-based dosing schemes in NSCLC (47) and melanoma (60). Exposure-response modeling in melanoma for nivolumab at doses of 0.1–10 mg/kg every two weeks has shown linear pharmacokinetics with time-varying clearance and without exposure being a significant predictor of response or survival. The FDA has since recommended a flat-dosing scheme for both nivolumab and pembrolizumab.

PD-L1 antibodies have been found to more effectively block PD-1 signaling in vivo (61) and have less PD-L2 interaction (62). This difference has led some to propose differential efficacy and adverse events, although prospective head-to-head comparisons have not been made. A meta-analysis of 11,379 clinical trial patients found improved survival and PFS with anti–PD-1 inhibitors compared with anti–PD-L1, whereas adverse events were similar (63). PD-1 inhibitors have been associated with increased risk of grade 3 to 4 colitis (0.85% vs. 0.34%, relative risk; 2.52) compared with PD-L1 inhibitors (64).

In preclinical studies, the combination of anti-CTLA4 and anti–PD-1 results in an approximate doubling of the tumor rejection rate compared with anti–PD-1 alone. In the tumor microenvironment of immune-checkpoint inhibitor combination-treated tumors, increased CD8+ cytotoxic T-cell and CD4+ T effector (Teff) cell tumor infiltration can be observed, along with reduced immunosuppressive T regulatory cells (Treg) and myeloid-derived suppressor cells (MDSC; refs. 65, 66). As discussed, this combination regimen has led to improved response rates and OS in metastatic melanoma, dMMR colorectal cancer, and HCC.

Expanding Dual Checkpoint Blockade Approaches

Building on the principle of dual checkpoint inhibition with anti-CTLA4 and anti–PD-1/PD-L1 to enhance responses, targeting other checkpoints in combination with PD-1/PD-L1 inhibition has been an area of intense investigation with the aim of improving responses and optimizing toxicity rates. The immune checkpoint indoleamine 2,3-dioxygenase 1 (IDO1) results in suppression of Teff and natural killer (NK) cells in the tumor microenvironment, and increases Treg and MDSC activity, leading to immunosuppression (67, 68). The combination of anti-CTLA4 and IDO1 inhibition in mice bearing B16 melanoma resulted in improved survival compared with anti-CTLA4 alone; however, single-agent IDO1 inhibition did not demonstrate antitumor effects in vivo (69). Similarly, in clinical trials assessing IDO1 inhibitors, single-agent therapy led to limited or no responses (70, 71). A number of phase II studies of IDO1 inhibitors in combination with anti–PD-1 therapy across solid tumors demonstrated promising response rates and reduced toxicities (13%–24% grade 3–4 toxicity) compared with CTLA4 plus PD-1/PD-L1 inhibitor combinations (72). However, research enthusiasm was significantly reduced when a phase III trial in checkpoint-naïve metastatic melanoma demonstrated no PFS benefit of the IDO1 inhibitor (epacadostat) in combination with pembrolizumab versus the PD-1 alone (mPFS: 4.7 vs. 4.9 months; ref. 73). This study also highlighted the need for a better understanding of the underlying biology, and for improved biomarkers for therapeutic efficacy and patient selection.

T cells in a chronically exhausted state can coexpress multiple checkpoints, suggesting that the targeting of a number of these, concurrently or sequentially, may be required to invoke an antitumorigenic response (74). Studies have reported that the upregulation of the TIM3 immune checkpoint correlates with resistance to anti–PD-1 agents in both in vivo and clinical samples (75–77). Early results from the combination of anti-TIM3 anti–PD-1/PD-L1 therapies have been promising. The AMBER trial of a TIM3 inhibitor in combination with an anti–PD-1 inhibitor reported partial responses in 4 of 31 patients with advanced NSCLC who had previously progressed on PD-1/PD-L1 inhibitors (78). A trial of the anti-TIM3 agent LY3321367 given as monotherapy reported one partial response out of 23 in a patient who had previously progressed on CTLA4 plus PD-1 inhibitor combination therapy, with results from the combination of LY3321367 and the PD-L1 inhibitor LY3300054 awaited (79).

The immune checkpoint LAG3 mediates the suppressive activity of Tregs and regulates T-cell expansion and homeostasis, with blockade of LAG3 resulting in an antitumorigenic TH1 phenotype (80–82). In preclinical models, the combination of PD-1 and LAG3 blockade resulted in improved tumor rejection compared with PD-1 inhibition alone (83–85). A phase II combination trial of LAG525, targeting LAG3, and the PD-1 inhibitor spartalizumab in 121 heavily pretreated patients with a range of solid tumors demonstrated one complete response and 11 partial responses (9.9%) with combination therapy, although no responses were observed with single-agent LAG3 blockade (86). Another LAG3 inhibitor, relatlimab, in combination with nivolumab demonstrated a response rate of 11.5% in 61 patients with melanoma whose disease had progressed on prior anti–PD-1/PD-L1 therapy. This response rate appeared to be 3.5-fold higher in patients with LAG3 expression of at least 1% versus those with less than 1% LAG3 expression (87). This relatlimab combination is now being assessed in a phase II/III trial in patients with advanced melanoma in the first-line setting. In a smaller study of 15 patients with the anti-LAG3 agent MK-4280 with pembrolizumab, 4 of 15 patients achieved a partial response (88). The lack of single-agent LAG3 inhibitor activity has led to caution; however, there is a clear and urgent clinical need to understand mechanisms of resistance to current immunotherapy regimens so as to develop new options for patients who develop progressive disease on immunotherapy. The PLATforM study aims to address this critical question with the combination of LAG525 and spartalizumab, as well as spartalizumab in combination with c-MET or CDK4/6 inhibition or anti-IL1β in patients with advanced metastatic melanoma (89). This adaptive trial design represents a rational approach and may be a model for future clinical trials in the post-immunotherapy space.

Bispecific Antibody Development and Potential

The concept of dual checkpoint inhibition may lead to higher-order combinations and will be expanded upon later in this review. An emerging approach that promises further specificity in targeting multiple checkpoints is the development of bispecific antibodies (90). This evolving technology, which allows a monovalent antibody that can target two or more receptors, opens up multiple potential approaches to drug development in this space. Specifically, some of the first emerging bispecific antibodies are targeting two checkpoints such as bispecifics to PD-1 and CTLA4, allowing a more specific targeting of the CTLA4 receptor on PD-1–positive cells and promises to uncouple efficacy from toxicity of this combination. The interesting characteristic of this approach is that which receptor the bispecific has higher affinity to (e.g., PD-1) will define which cells will be targeted for inhibition of the second receptor (e.g., CTLA4), and sometimes at hundreds of fold higher affinity (ref. 91; NCT03517488).

From an immunotherapy resistance perspective, using bispecifics can allow targeting specific mechanisms of resistance such as TGFb or MET inhibition (92). For instance, a particularly interesting molecule that has a PD-L1 antibody and TGFβ-TRAP as the second part of the bispecific allows TGFβ levels—a well-described mediator of resistance in the tumor microenvironment—to be modulated favorably. This approach is currently in clinical testing alone or in combination (ref. 93; NCT 04349280). Virtually every mechanism of resistance described in this review could potentially be amenable to targeting by bispecific antibodies. Bispecifics also permit the targeting of specific cells as well. For example, T-cell engagers or BiTEs are bispecifics bearing CD3 motifs that can attach a T cell directly to a tumor cell and initiate an immune response that is MHC-independent (93). Trispecifics are also being developed with a focus on NK cells in so-called trispecific NK-cell engager therapeutics (TriNKET). Those novel agents are entering the therapeutic armamentarium and will likely be used alone or in combination with other therapeutics, including chemotherapy and TKIs, allowing multipronged targeting of immunotherapy resistance.

Neoadjuvant Combination Immune Blockade

Delivering combination immune-checkpoint inhibition in the neoadjuvant setting takes advantage of the in situ tumor to stimulate immune responses, resulting in increased clonal expansion of T cells compared with adjuvant IO (94). Systemic immunosuppression is observed in the metastatic setting (95), and patients with metastatic disease have often experienced a prior treatment course including immunosuppressive treatments. Therefore, it could be hypothesized that treating at the earliest stage possible not only results in increased exposure to tumor antigens, but also utilizes an active, responsive immune system that may improve long-term systemic immunity.

In keeping with this, the combination of nivolumab–ipilimumab results in improved pathologic complete response (pCR) rates in resectable melanoma compared with nivolumab alone (45% vs. 25% in a 23-patient study), although with an increased toxicity cost (73% grade 3–4 toxicities vs. 8%; ref. 96). A randomized trial examined three different doses and schedules of the combination of ipilimumab and nivolumab in the neoadjuvant therapy of melanoma and concluded that two doses of ipilimumab at 1 mg/kg and nivolumab at 3 mg/kg intravenously 3 weeks apart gave the optimal outcome of more than 70% pCR rate and 30% toxicity and is the currently accepted neoadjuvant regimen (97). Similarly, in resectable colorectal cancer, neoadjuvant nivolumab–ipilimumab for 4 weeks resulted in pathologic responses in 100% of MSI cancers, and 27% of MSS disease (98). Grade 3 to 4 toxicities were experienced by 13% with one patient (out of 40) requiring infliximab for checkpoint-induced colitis. In head and neck cancer, where palliative immunotherapy has been effective, neoadjuvant nivolumab–ipilimumab resulted in responses in 69% of patients, with limited toxicity from this short course of treatment (99). Neoadjuvant combinations with chemotherapy or other standard-of-care agents are also promising, with combinations in breast cancer discussed below.

Exploiting Costimulatory Molecules to Enhance Antitumorigenic Activity

Activating the immune system using costimulatory molecules in combination with anti–PD-1/PD-L1 has been shown to lead to improved outcomes by enhancing antitumorigenic activity. For example, activation of IL2 signaling via CD122 results in activation of naïve CD8+ T cells and also increases NK-cell activity (100, 101). The combination of the CD122 agonist NKTR-214 and anti–PD-1 demonstrated promising results in preclinical models of NSCLC and colorectal cancer (102). In treatment-naïve metastatic melanoma, the combination of NKTR-214 and nivolumab resulted in an ORR of 59.5% (22/37), with 7 (18.9%) patients experiencing a complete response (103). This combination has now been granted the breakthrough therapy designation from the FDA, which would expedite this combination through ongoing phase III studies. Other costimulatory molecules, such as 4-1BB, OX-40, and GITR, similarly result in increased CD8+ and NK-cell activity and reduced Treg-mediated immunosuppression, with multiple phase I/II trials ongoing in combination with immune-checkpoint blockade (104–106).

Innate immune-stimulating agents, such as TLR9 or stimulator of interferon genes (STING) agonists, delivered intratumorally can prime the tumor microenvironment prior to immune-checkpoint therapy. A number of TLR9 agonists (CpG-oligodeoxynucleotides) are in ongoing phase I to III studies in combination with anti–PD-1 and anti–CTLA4 immune-checkpoint inhibition. A phase Ib study of the TLR9 agonist SD-101 in combination with pembrolizumab resulted in an ORR of 78% in IO-naïve patients with melanoma and 15% of checkpoint (anti–PD-1) refractory disease (107), with CD8+ and NK-cell trafficking to the tumor site. A similar approach with lefitolimod and ipilimumab is ongoing in an all-comer study (108). Following promising results in phase II, with 38% ORR in anti–PD-1–refractory melanoma (109), the combination of tilsotolimod and ipilimumab is now under investigation in the phase III study ILLUMINATE301 (110). Initial studies of intratumoral STING agonists have also demonstrated the ability of these agents to activate local immune responses and synergize with anti–PD-1 therapy (111, 112), although mixed results from trials to date have demonstrated the need for patient selection for these approaches, and further preclinical studies to understand potential resistance mechanisms.

Immune-Checkpoint Inhibition and CAR-T Cell Therapy

Chimeric antigen receptor (CAR) T cells directly target tumor-specific antigens, independent of MHC expression, with successful outcomes in a number of hematologic malignancies (113). However, replicating this success in solid tumors has proved challenging. One potential resistance mechanism is the upregulation of immune-checkpoint expression common to solid tumors, causing CAR-T cells to become exhausted and ineffective. In mesothelioma, which has already demonstrated positive efficacy data with nivolumab–ipilimumab combination therapy (114), CAR-T cells targeting mesothelin followed by anti–PD-1 resulted in an ORR of 50% (7/14 patients; ref. 115). CAR-T cells can also be manipulated using CRISPR/Cas9 to disrupt PD-1 expression. The advantages of this approach include a potential reduction in systemic effects of PD-1 inhibition, with highly specific PD-1 knockdown only in tumor-targeted T cells, and avoiding repeated administrations of systemic anti–PD-1 (116). Both PD-1–disrupted CAR-T cell therapy and combinations with systemic anti–PD-1 therapy are the subject of an ongoing study in the early-phase setting (117).

Improving Patient Outcomes with Chemotherapy and Radiotherapy Combinations

The role of the immune system in response to chemotherapy has been noted since the 1970s, when studies assessing the mechanisms of action of anthracyclines demonstrated an improved response in immunocompetent murine models (118). Immunogenic cell death (ICD) has been proposed as a key mechanism resulting in an immune-infiltrated tumor microenvironment favorable for immunotherapy, suggesting that ICD-inducing regimens may synergize with immune-checkpoint inhibitors (119). Ongoing chemotherapy–immunotherapy combination trials are discussed in greater detail in other review articles (120). The combination of platinum chemotherapy and immune-checkpoint blockade has been successful in SCLC (22), with the immunogenic response generated by chemotherapy overcoming the lower response rates observed with anti–PD-1 treatment alone in low PD-L1–expressing tumors (31). DNA-damaging chemotherapy additionally activates the innate immune cGAS–STING pathway via cytosolic DNA, resulting in an immune-rich microenvironment that may predict responses to checkpoint blockade (121–123).

Although taxanes do not directly cause ICD, they decrease intratumoral immunosuppressive MDSCs and Tregs, augmenting cytotoxic T-lymphocyte activity (124, 125). The combination of nab-paclitaxel and atezolizumab is approved for PD-L1–positive metastatic TNBC, but was not effective in the PD-L1–negative population (126). Alternative chemotherapy regimens may further improve patient outcomes, as illustrated by the ISPY-2 studies in neoadjuvant breast cancer with pembrolizumab and standard anthracycline-based neoadjuvant chemotherapy (127). In these studies, neoadjuvant pembrolizumab and paclitaxel followed by anthracycline–cyclophosphamide resulted in pathologic complete response (pCR) rates of 60% in TNBC compared with 22% without pembrolizumab, and 30% versus 13% in hormone receptor–positive/HER2-negative disease (128). In a similar setting, four cycles of pembrolizumab in combination with paclitaxel–carboplatin chemotherapy, followed by anthracycline—cyclophosphamide, in TNBC resulted in a pCR of 64.8% versus 51.2% without pembrolizumab. In this study, patients received adjuvant pembrolizumab or placebo for a further nine cycles following resection, with follow-up ongoing (129).

DNA damage by radiotherapy is associated with a range of immunogenic effects, including activation of the cGAS–STING pathway, increasing neoantigen expression, and upregulation of PD-L1 expression (130–132). Sequential chemoradiotherapy–IO has been shown to lead to significantly improved patient outcomes in stage III NSCLC (27, 28), whereas a phase II study of concurrent anti–CTLA4 plus radiotherapy in NSCLC suggested improved responses in patients previously resistant to anti–CTLA4 given alone or in combination with chemotherapy (133). Although more than 100 trials of anti–PD-1/PD-L1 therapy in combination with chemo/radiotherapy are ongoing (134), questions remain over the optimal dose and schedule of radiotherapy–immunotherapy combinations. For example, serial low doses of radiation have been shown to activate a STING-mediated immune response, whereas higher doses (i.e., ≥12–18 Gy) instead activate TREX1, removing cytosolic DNA and preventing STING activation (135). Concurrent radiation may result in enhanced responses, but this may be dependent on the target (CTLA4 vs. PD-1/PD-L1), dose, and delivery of the radiation using proton beam or other approaches (136). Although the abscopal effect of radiotherapy may be enhanced by immune-targeting therapies, clinical evidence for this approach has thus far been largely confined to case reports rather than large-scale trials (137, 138).

Activating Innate Immunity with DNA Damage Response Inhibitors

DNA damage response (DDR) deficiency results in activation of the innate immune system via the critical cGAS–STING pathway, which is required for interferon expression in response to cytoplasmic DNA (124, 139, 140). A number of studies have now demonstrated cytoplasmic DNA and tumor cell–intrinsic activation of the STING pathway following treatment with agents targeting the DDR, in particular PARP inhibitors (141–144). PARP inhibition leads to activation of intratumoral dendritic cells and increased CD8+ T-cell infiltration via activation of the STING pathway. Interestingly, PARP inhibition also reduces the PARylation of STAT3, while increasing STAT3 transcriptional activity and expression of PD-L1 (145). These studies suggest that PARP inhibition acts synergistically with immune-checkpoint blockade in both DNA repair–deficient (e.g., BRCA1/2-mutant) and DNA repair–proficient cancers (146).

Early-phase trials of PARP inhibitor plus immune-checkpoint inhibitor combinations have been promising, with activity demonstrated in patients with advanced castration-resistant prostate cancer (147, 148), TNBC (149), and ovarian cancer (150). This combination has demonstrated antitumor activity in BRCA1/2 wild-type TNBC and platinum-resistant ovarian cancer, populations that are typically resistant to single-agent PARP inhibitors. Importantly, bone marrow suppression typical of PARP inhibitor toxicity was shown to occur at similar rates to that observed with single-agent therapy, and no significant increase was found in immune-related toxicities.

Targeting the ATR/CHK1 pathway also results in activation of a STING-dependent immune response (143), and ATR inhibitors have demonstrated promising clinical activity in combination with durvalumab (151). There is also growing evidence for a number of DDR inhibitors in combination with immune-checkpoint inhibitors, including those targeting WEE1 (152), ATM (153, 154), and DNA-PK (155). With improved understanding of the impact of these novel DDR targeting agents on the immune response in specific patient subgroups, it is tempting to propose that future DDR inhibitor combination therapies will focus on targeting specific patient populations, based on the DNA-repair capacity identified in individual tumors, in combination with immune-targeting agents designed to appropriately modulate the tumor microenvironment.

Increasing Tumor Immunogenicity with Molecularly Targeted Agents

A number of targeted therapies are recognized to increase the immunogenicity of tumors and are therefore predicted to work synergistically with immune-checkpoint blockade. For example, inhibitors of the MAPK pathway result in increased expression of MHC I on the tumor cell surface, with subsequent increase in antigen-specific T effector cells and T cell–mediated cytotoxicity (121, 156). This combination has been hampered in early clinical trials by associated increases in toxicity rates, with a trial of anti-CTLA4 and the BRAF inhibitor vemurafenib terminated due to a high rate of hepatotoxicity (157). The addition of the MEK inhibitor trametinib, while reducing hepatotoxicity, resulted in colitis and bowel perforation in two of seven patients receiving this triplet combination (158). More promisingly, a phase II study of BRAF and MEK inhibition with anti–PD-1/PD-L1 therapy in treatment-naïve BRAF-mutant melanoma reported tolerability (58% grade 3–5 toxicities vs. 27% in patients receiving BRAF and MEK inhibition alone) and improved PFS (12 month PFS 59% vs. 45%; ref. 159). A phase III randomized trial in patients with unresectable stage IIIC to IV BRAFV600-mutant melanoma assessed vemurafenib plus cobimetinib with atezolizumab versus placebo with atezolizumab. In the triplet therapy group, PFS was superior versus the reference arm (15.1 vs. 10.6 months; HR 0.78; 95% CI, 0.63–0.97; P = 0.025), with minimal increases in grade 3 to 4 toxicities (79% vs. 73%), and this study resulted in FDA approval of the triplet combination in the first-line therapy of BRAF-mutant metastatic melanoma (160). However, a phase III study of MEK inhibition with atezolizumab in heavily pretreated MSS colorectal cancer did not result in improved survival compared with standard-of-care regorafenib therapy alone (161).

Epigenetic modification by histone deacetylase (HDAC) inhibitors reduces the number of MDSCs in the tumor microenvironment in preclinical models, resulting in improved response to anti–PD-1 blockade (162, 163). In addition, treatment with HDAC inhibition leads to upregulation of the chemokines CXCL10 and CCL5 in the tumor microenvironment that, in turn, drives recruitment of antitumorigenic CD8+ T cells (164). A phase Ib/II trial of the HDAC inhibitor entinostat with pembrolizumab in patients with metastatic melanoma who had previously progressed on immune-checkpoint therapy demonstrated responses in 10 (18.9%) of 53 patients, including one complete response (165). Treatment with demethylating agents such as azacitdine upregulates MHC I on tumor cells and also unmasks endogenous retroviruses, resulting in cytoplasmic double-stranded RNA (dsRNA) that activates innate immune responses, increasing tumor immunogenicity (37, 166, 167). A combination study of azacitidine and nivolumab demonstrated responses in relapsed or refractory acute myeloid leukemia (168). However, a phase II study of azacitidine and pembrolizumab in MSS colorectal cancer demonstrated only one partial response out of 31 patients to the combination therapy (169).

Given our current superficial knowledge of the therapeutic effects of different combinations on the immune system, multiple preclinical tumor models have been developed to provide a deeper understanding of the mechanisms of action and to help predict the efficacy and safety of different immunotherapeutic interventions in patients with cancer. As summarized in Table 2, many proof-of-principle preclinical studies involving a wide range of immunotherapy combinations have been conducted with different murine models. The results from these studies provide a compendium of immunotherapy combinations for future clinical assessment. Although none of these murine models completely mirror the heterogeneity and adaptability of the antitumor immune network in patients with cancer, each one has its unique value in the cancer immunotherapy research field. Therefore, understanding the pros and cons of each model can guide the optimal selection of appropriate murine tumor models for preclinical assessment of different immunotherapy combinations. Current murine tumor models may be summarized into three categories based on the types of immune systems involved: syngeneic, chimeric, and humanized tumor models.

Syngeneic Tumor Models

Mouse models in this category are fully immunocompetent. Tumor development in these mice is induced either by inoculation of murine cell lines or by tissue-specific activation of oncogenic pathways. Due to the general ease of use, inoculating wild-type mice with established syngeneic tumor cell lines is by far the most commonly used approach to generate syngeneic tumor models. Several inbred strains such as C57BL/6, BALB/c, 129/sv, and DBA/2 have been used to generate a variety of murine cell lines to represent different cancer types, including but not limited to B16 (melanoma; ref. 170), MCA205 (sarcoma; ref. 171), A20 (lymphoma; ref. 172), P815 (mastocytoma; ref. 173), 4T1 (breast cancer; ref. 174), 344SQ (lung cancer; ref. 175), ID8 (ovarian cancer; ref. 176), MB49 (bladder cancer; ref. 177), MC38 (colon cancer; ref. 178), and CT26 (colon cancer; ref. 179). The majority of these tumor cell lines display a certain degree of genome instability, which results in the expression of tumor-specific antigens that may be recognized by the adaptive immune system, particularly T cells (180, 181). Because mice challenged with these tumor cells have intact murine immune systems, a “physiologically relevant” tumor immune microenvironment can be achieved in these tumor-bearing mice.

However, host mice used in syngeneic cell line models are commonly young and healthy inbred mice, which are housed in specific pathogen-free conditions. In addition, there is mounting evidence supporting a high divergence in immune responses among inbred mouse strains and cell lines. For example, the C57BL/6 strain is TH1-biased, whereas BALB/c and DBA/2 strains are TH2-biased (182). Discrepancies in the antitumor effects of IO combinations may thus be observed depending on the murine tumor cell line model chosen. Therefore, to increase the likelihood of successful clinical translation, the use of multiple murine tumor cell line models on a range of genetic backgrounds is encouraged. Moreover, introducing fundamental cancer-driven mutations into tumor cell lines or perturbing environmental factors in housing conditions, such as diet and microbiome, should be used to more broadly recapitulate interpatient heterogeneity.

Recent advances in technologies of in vivo genetic manipulation allow us to precisely control the timing, duration, and tissue type of gene expression. Using these approaches, genetically engineered mouse models (GEMM) have been applied in immunotherapy studies to better mimic the heterogeneity of the natural steps of tumor development in patients with cancer. The Mouse Tumor Biology Database (http://tumor.informatics.jax.org) provides a comprehensive list of GEMMs for a variety of human cancer types (183). As tumor development in GEMMs is driven mainly by one or two genetic alterations in dominant oncogenes, these models generally have a low tumor mutation burden. Furthermore, de novo tumor growth in GEMMs provides sufficient time to trigger a broad range of immunosuppressive mechanisms within the tumor microenvironment. This lack of neoantigens and the profound immunosuppression make GEMMs typically highly resistant to cancer immunotherapy, sometimes even more so than clinically equivalent malignancies. For example, melanomas derived from the Tyr gene promoted BRAF-mutant PTEN-loss model (184) have been demonstrated to be resistant to immune-checkpoint inhibitors (185). Depleting Tregs also failed to control the development of tumors with transgenic expression of the RET receptor tyrosine kinase gene, suggesting that other immunosuppressive cells play an important role and replace immunosuppressive, tumor-promoting functions of Tregs (186). Therefore, effective immunotherapy strategies should consider including the inhibition of Treg migration into the tumor combined with neutralization of other immunosuppressive cells and factors in the tumor microenvironment (186). These models have now been used to address the challenges identified in patients with cancer with low immunogenic tumors or immune-resistant tumors. Peng and colleagues utilized the BRAF-mutant PTEN-loss GEMM to demonstrate that inhibition of the oncogenic activation of the PI3K pathway by PTEN loss can sensitize tumors to immune-checkpoint blockade therapy (187). Despite the expensive and time-consuming procedures required, GEMMs are currently the optimal models to use to evaluate the potential of IO combinations to overcome immune resistance, particularly resistance associated with genetic alterations.

Chimeric Tumor Models

Although syngeneic murine models provide an opportunity to characterize the immunologic changes associated with IO combinations in the presence of an intact immune system, they cannot be used to test the human version of IO reagents due to cross-species differences. To circumvent this limitation, chimeric murine tumor models have been developed by knocking in human immune-related genes. In these immunocompetent tumor models, the majority of immune compartments are murine-based, whereas the knocked-in human genes allow a portion of their immune system to be controlled by certain human immune factors. The first human knock-in model was the HLA-A2 murine strain. Mice carrying a human MHC class I gene (HLA-A2.1) were created to evaluate the antitumor activity of HLA-restricted vaccination strategies (188). Since then, further models have been generated to study clinical-grade immunomodulatory agents, including anti-human CTLA4, OX40, or Tim3 (189–191). These studies successfully confirmed the in vivo efficacy of immunomodulatory agents alone or in combination with other treatment modalities. Chimeric models are, therefore, valuable in validating combination effects of clinical-grade reagents targeting novel immune regulators in vivo, with commercial services now available to generate customized models.

Humanized Tumor Models

To better represent tumor heterogeneity within patients with cancer, efforts have been made to generate humanized tumor models. In these models, immunocompromised mice are partially reconstituted with the human immune system by transplantation of CD34+ hematopoietic progenitor cells (HPC) from human umbilical cord blood, bone marrow, or peripheral blood. The implantation of tumor tissues or cell lines derived from patients with cancer in humanized host mice is used to induce tumor development. The NSG strain, which carries the NOD/SCID IL2Rγ chain knockout and lacks functional T, B, and NK cells, is most frequently used as the host strain for humanized models. Additional genetic modifications have recently been performed in NSG and other immunodeficient mice to express human cytokines, including stem cell factor, M-CSF, GM-CSF, and/or IL3, which are essential for the growth and differentiation of HPCs and to support human myeloid reconstitution (192, 193). The expression of human cytokines in these new strains, namely NSG-SGM3, NOG-ExL, MSTRG, and MISTRG, significantly increases the engraftment rate of the human immune system (194). The efficacy of pembrolizumab in a CD8+ T cell–dependent manner was successfully reproduced in these humanized models (195, 196).

However, antitumor immune responses in these models come largely from allogenic immune cells. Compared with natural tumor rejection through tumor-associated antigens, the antitumor immune responses mediated by allogenic rejection may be more intense. Moreover, the cost of humanized tumor models is dramatically higher than the other two types of models described above. Therefore, humanized tumor models are more suited for validating the efficacy and safety of the human versions of immunotherapy combinations than discovering novel combinations.

Highly active antiretroviral therapy (HAART) consisting of four-drug combinations that completely suppress human immunodeficiency virus (HIV) replication has evolved to become so efficacious yet tolerable that it can even be used as a routine prophylactic in “at risk” populations. HIV therapy began, however, with single, toxic drugs with limited efficacy, which gained efficacy but even greater toxicity in two-drug combinations. It was only with the inception of truly virus-specific therapeutics that high-order, high-efficacy combination therapies became practical. In cancer, high-order (three or more) immunotherapies have now begun to enter the clinic with promising initial signs against “cold” tumors. Much like HAART, however, the development of widely applicable and broadly efficacious multiple IO combinations will necessitate the development of more tumor-specific modulators of host immunity.

Due to the toxicities inherent in high-order combinations of existing immunotherapies, these trials have thus far been limited to patients with metastatic “cold” cancers (Fig. 1). In preclinical models of pancreatic ductal adenocarcinoma (PDAC), for example, only high-order combinations of chemotherapy, T-cell checkpoint blockade, and antigen-presenting cell activation via CD40 agonism could promote durable benefit and tumor regression (197, 198). This combination of checkpoint blockade, CD40 agonist antibody, and chemotherapy has recently reported promising phase I clinical trial data in PDAC, and final data are eagerly awaited (199). In this case, existing agents were combined successfully through thoughtful sequencing in order to avoid intolerable levels of additive toxicity.

In many cases, the ideal setting for high-order IO combinations would necessitate neither dose reduction to suboptimal levels nor sequencing of component therapies. To this end, multiple immunomodulatory antibodies have entered the clinic having been engineered to act selectively within the tumor microenvironment, thus sparing the host from adverse events. These novel approaches can be broadly classified into conditionally active versus bispecific antibodies. The most advanced example of conditionally active antibodies are “probodies,” in which the antigen-binding region of the antibody is blocked by a peptide that is tethered to the antibody with a linker containing multiple cleavage sites for tumor-selective proteases (200). In a phase I trial of the PD-L1 probody CX-072, only 6% of the 72 patients treated experienced a grade 3 or greater adverse event and no adverse event–related discontinuations occurred (201). The alternative approaches to tumor selectivity are bispecific antibodies in which one arm of the antibody, usually the higher affinity, binds to a tumor or tumor microenvironment selective antigen, thus localizing the effect of the lower affinity active arm. The CD137 (4-1BB) and HER2 bispecific antibody PRS-343, in which 4-1BB activation is sequestered in the tumor microenvironment by HER2 binding, is one of the most advanced examples of this approach (202). Although CD137 activation can cause liver toxicity in certain patients (4), this construct safely sequesters its activity to the tumor. PRS-343 was safe and well tolerated in the first-in-human phase I trial involving 53 HER2+ patients with advanced solid tumors. Two patients achieved radiologic responses, whereas other patients had stable disease. Importantly, PRS-343 demonstrated a potent increase in CD8+ T-cell numbers and proliferative index in the tumor microenvironment of responders, suggestive of 4-1BB agonism on T cells. By localizing the effect of checkpoint blocking and costimulatory agonist antibodies to the tumor, these approaches set the stage for tolerable combinations of multiple synergistic immunotherapies, for example, CTLA4, PD-1, and 4-1BB inhibitor combination therapy.

An alternative emergent approach to multimodal immunotherapy circumvents systemic toxicity concerns by administering some or all therapeutics directly into the tumor itself. Innate immune agonists such as activators of Toll-like receptors or the STING pathway are injected intratumorally in a single lesion to generate T-cell responses, which can then traffic to and eradicate distal sites of cancer (abscopal effect; ref. 203). Although clinical combinations with these therapies pair them with a single systemic checkpoint antibody, preclinical studies have demonstrated that as many as three immunomodulatory antibodies can be coinjected with a STING agonist, resulting in superior capacity to eliminate noninjected sites of disease (204, 205). With this type of nontoxic scaffold to build upon, current efforts can focus on identifying systemic therapies to weaken the immune resistance of uninjected sites of cancer, making it easier for the injection-mobilized T cells to eradicate them.

Although novel, less toxic immunotherapies will form the backbone of most emergent high-order IO combinations, the effect of these therapies may be augmented and extended through the addition of FDA-approved, even over-the-counter, drugs that can contribute to antitumor immunity without significant toxicities. Extracellular adenosine, for example, plays a clearly established role in immune suppression in the tumor microenvironment (206). Although still pending approval in the United States, the adenosine receptor blocking compound istradefylline is approved in Japan for the treatment of Parkinson's disease, where it has shown high tolerability (207). Such adenosine receptor antagonists may potentially be a promising addition to any immunotherapy combination (208). Multiple studies have demonstrated antitumor efficacy of inhibition of the renin–angiotensin system through decreased suppressive polarization and activation of tumor stroma (209). These drugs are commonly used to treat cardiovascular disease and would be a rational addition to immunotherapy combination regimens. Platelets have also been implicated in tumor immune suppression, which can be blocked by clopidogrel and over-the-counter aspirin (210). PDE5 inhibitors may also have value through their capacity to inhibit MDSCs (211). Further study is warranted to assess the capacity of these nontoxic, widely available drugs to augment tumor immunity; however, one or more could likely become key components of multimodal immunotherapy combination therapy.

Finally, the greatest challenge to development of these high-order combinations may not be toxicity or engineering, but rather finding a system in which the most effective combinations can be screened, so that only the most promising ones enter clinical trials. As discussed above, there are more than 2,250 trials evaluating various PD-1/L1 inhibitor combinations, a number that is increasingly straining resources and the availability of appropriate patients to evaluate potential immunotherapies. The number of possible permutations involved in developing five-drug IO combination therapies makes primary screening in patients impractical, if not impossible. In this setting, drugs must first be prioritized based on single-agent efficacy, underlying biological rationale for combination, and lack of additive toxicity. Potential combinations involving such agents can then be screened through appropriate murine tumor models of cancer to identify the most promising candidates for study in patient trials. Using such a defined development and screening approach and leveraging existing therapies, novel engineering, and selective FDA-approved drugs may provide a path to multimodal IO combinations that approach the efficacy and tolerability of HAART regimens for HIV.

Drug development to reverse resistance to immunotherapy needs to take into account multiple streams of knowledge being generated in the preclinical, translational, and clinical spaces. For instance, PD-L1 testing seems to predict a better response to immunotherapy across the board. However, due to its poor ability to discriminate antitumor efficacy in cancers such as melanoma, it has not entered clinical practice as a useful test. However, given our increasing abilities to pathologically, genomically, and transcriptomally characterize cancers, we have a mandate to now hone drug development into better-defined patient populations where therapeutic benefit can be augmented.

A recent effort initiated by the Society for Immunotherapy of Cancer (SITC) provided updated and improved clinical definitions of drug resistance to harmonize and standardize drug development efforts in the PD-1 inhibitor resistance space (212). Specifically, the SITC Resistance Taskforce defined three main clinical patterns of resistance to single-agent anti–PD-L1 antibodies: (i) Primary resistance that occurs while on treatment with single-agent therapy; this pattern requires patients to have adequate exposure to the agent (at least six weeks), to have shown no evidence of therapeutic benefit [no complete response (CR) or partial response (PR) or stable disease (SD) > 6 months], and to have a confirmatory scan to confirm progression (and predominantly to rule out pseudoprogression). (ii) Secondary resistance is defined as tumor progression after an initial clinical benefit (achievement of radiologic CR, PR, or SD > 6 months), and also requires a confirmatory scan. (iii) The third pattern defined in this effort is recurrence or progression of the tumor after halting therapy. This definition assigns the pattern to be similar to either primary or secondary resistance depending on the setting of therapy (adjuvant, neoadjuvant, or advanced disease) and the time from when therapy was discontinued (<12 weeks or >6 months).

Another clinical pattern of resistance is site-specific resistance, where patients may experience control of the disease in most of their body but have progressive lesions in one or two organs. The central nervous system (CNS) is a particularly relevant location and has emerged as a major therapeutic challenge. For instance, in melanoma, it has been reported that the CNS is the most frequent site of first progression on single-agent checkpoint blockade (213). Moving forward, it will be important to include this population of patients with progressing brain metastases in trials of novel agents, so as to enhance our understanding of the intracranial activity of immunotherapeutic agents, as well as to develop more brain-penetrant agents and approaches that tackle this most devastating complication of cancer progression. This has been recognized by the FDA, which has issued guidance to increase the representation of patients with CNS metastases on clinical trials (214).

Improved patient selection on immunotherapy combination clinical trials may seem like an aspirational goal at this time, but all the elements now appear to be falling into place, and a concerted effort between academia and industry partners could indeed accelerate this approach to achieve improved patient outcomes. Some of the examples listed above provide context to this evolving immunotherapeutic landscape. For instance, the failure of the IDO inhibitor epacadostat in a large phase III melanoma study, despite demonstrating somewhat promising data in early-phase trials, has been attributed largely to the fact that this was an unselected population, and this agent may indeed still have useful activity in a subset of patients (73, 215, 216). It is therefore encouraging to see how the development of other immunotherapy combination trials seems to be informed by that experience. An example is the anti-LAG3 antibody relatlimab and nivolumab combination already described above. The phase I data of this combination in the second-line setting indicated an ORR of 11% in unselected patients with melanoma, but was 18% in LAG3-positive patients (87). Subsequently, a phase II/III trial of relatlimab was initiated in the first-line setting in melanoma, which, although all first-line patients are still eligible, does require screening of patient tumor samples obtained within a three-month period before enrollment and for evaluable results from PD-L1 and LAG3 IHC staining to be available prior to patients being randomized (NCT03743766). Additionally, the study is stratified for both of these biomarkers, as well as BRAF mutations, which will be incredibly valuable at the time of study analysis because one can start gleaning a specific effect of the combination in a particular subset of patients. In general, such biomarker-driven approaches will of course have to be carefully designed by taking into account all the statistical caveats of subgroup analyses.

This trend is now becoming increasingly visible across the immunotherapeutic landscape, and still needs to be furthered, perhaps even more so in the immunotherapy-refractory setting in cancers where immune-checkpoint blockade has become a first-line approach. Translational and preclinical studies are continually identifying novel mechanisms of resistance to different immunotherapy agents on an ongoing basis. Investigators and pharma should be emboldened to conduct studies in molecularly characterized populations of patients where the agent in use specifically targets the biomarker used for patient selection. An example of such a strategy is the use of selective PI3Kβ inhibitors in patients harboring tumors with PTEN loss. Such a thoughtful biomarker-driven approach takes into consideration that certain key signaling networks that can be potently targeted in tumors, such as the PI3K pathway, are also critical for T-cell activation and function. Being able to identify specific molecularly targeted agents that may have a differential impact on tumor versus T cells is the new quest for a “therapeutic window.” This is especially true of novel metabolism-directed agents, such as glutaminase inhibitors (217).

There are also opportunities for the development of agents with pleotropic effects where patient selection may be challenging but have the potential to affect multiple nodes of immune activation and tolerance. Examples of those approaches are agents targeting epigenetic modulation, such as hypomethylating agents (218), HDAC inhibitors (219), and bromodomain and extraterminal domain (BET) inhibitors (220), which have genome-wide effects and affect both tumor cells and T cells.

In 2017, the FDA approved pembrolizumab for the treatment of MSI-H or dMMR regardless of primary tumor site (221), which has subsequently been supported by a randomized trial of pembrolizumab utilized for 27 different tumor types (222). However, MSI-H may have been a surrogate for high tumor mutation burden (TMB-high; ref. 223), which has subsequently been used as a biomarker for selection in both tissue- and blood-based whole-exome sequencing (224). A meta-regression analysis of 117 clinical trials for response in the TMB-high group, defined as ≥10 mutations per megabase, found single-agent PD-L1, CTLA4, and combination to significantly affect ORR (225). Prospectively, the multicohort phase II KEYNOTE-158 found an improved overall response rate of pembrolizumab in the TMB-high group compared with the TMB-low group (29% vs. 6%), leading to approval of single-agent IO for the TMB-high group (226).

In addition, there is an increasing appreciation of the role of host factors on the immune response unrelated to the intrinsic biology of tumor development and resistance. For instance, as gender and diet differences have long been observed to affect patient outcomes to cancer therapy, the microbiome has emerged as a critical element in modulating the immune response. There have been many high-impact publications indicating that the diversity and composition of the gut microbiome can indeed predict for higher response rates and better outcomes to therapy. Functional studies, as well as in vivo data, offer solid evidence that this is a causal effect resulting from the intricate interaction of gut bacteria with the immune system (227, 228). This has emerged as a novel therapeutic target, and multiple studies currently under way, in which modulation of the microbiome is being utilized with fecal microbiota transplantation, are showing early evidence of activity, including in patients with PD-1–resistant disease (229). Other interesting approaches utilizing diet to modulate the microbiome favorably are also under way.

Biomarker development continues to evolve at a rapid pace, and emphasis should continue to be placed on the incorporation of tumor biopsy and blood collection from patients on clinical trials, especially at the time of disease progression for the assessment of predictive biomarkers of resistance. This could yield a deeper understanding on how the tumor has evolved clonally and perhaps activated other pathways that could then be subsequently targeted. Although there are of course logistic, and occasionally even ethical, challenges, the data obtained from such clinical trial biopsies may also be incredibly valuable in identifying areas where the studied agent has failed in exerting its effects, offering potential pharmacokinetic and pharmacodynamic strategies to improve pharmacologic outcomes. Comprehensive and longitudinal profiling along the patient journey, within and outside a clinical trial, should become the norm rather than the exception.

T.A. Yap reports grants from Artios, Constellation, Cyteir, and Eli Lilly, Forbius, GlaxoSmithKline, Genentech, ImmuneSensor, Ipsen, Jounce, Karyopharm, Kyowa, Novartis, Ribon Therapeutics, Regeneron, Sanofi, Scholar Rock, Tesaro, and Vertex Pharmaceuticals, grants and personal fees from AstraZeneca, Bayer, Clovis, EMD Serono, F-Star, Merck, Pfizer, Repare, and Seattle Genetics, personal fees from Almac, Aduro, Atrin, Axiom, Bristol Myers Squibb, Calithera, Cybrexa, Guidepoint, Ignyta, I-Mab, Janssen, Roche, Rubius, Schrodinger, Varian, and Zai Labs outside the submitted work. E.E. Parkes reports personal fees from Boehringer Ingelheim outside the submitted work. J.T. Moyers reports nonfinancial support from Astellas Pharmaceuticals outside the submitted work. M.A. Curran reports personal fees from ImmunoGenesis, Inc., Agenus, Inc., Alligator Bioscience, Inc., Ammunix, Inc., Aptevo, Inc., and ImmunOs, Inc., Oncoresponse, Inc., Pieris, Inc., Nurix, Inc., Servier, Inc., Kineta, Inc., Xencor, Inc., and Salarius, Inc., and grants and personal fees from ImmunoMet, Inc., during the conduct of the study; in addition, M.A. Curran has a patent for antibodies that bind PD-L1 and prevent its binding to PD-1 pending and licensed to ImmunoGenesis, Inc., a patent for dual specificity antibodies which bind both PD-L1 and PD-L2 and prevent their binding to PD-1 pending and licensed to ImmunoGenesis, Inc., a patent for antibodies which bind PD-L2 and prevent its binding to PD-1 pending and licensed to ImmunoGenesis, Inc., and a patent for methods and composition for localized secretion of anti-CTLA4 antibodies issued and licensed to multiple licensees. H.A. Tawbi reports grants and personal fees from Bristol Myers Squibb, Merck, Novartis, and Genentech, grants from GSK, Celgene, and personal fees from Eisai and Iovance outside the submitted work. No other disclosures were reported.

1.
Chen
DS
,
Mellman
I
. 
Oncology meets immunology: the cancer-immunity cycle
.
Immunity
2013
;
39
:
1
10
.
2.
Sharma
P
,
Allison
JP
. 
Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential
.
Cell
2015
;
161
:
205
14
.
3.
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
.
4.
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
.
5.
Ascierto
PA
,
Del Vecchio
M
,
Robert
C
,
Mackiewicz
A
,
Chiarion-Sileni
V
,
Arance
A
, et al
Ipilimumab 10 mg/kg versus ipilimumab 3 mg/kg in patients with unresectable or metastatic melanoma: a randomised, double-blind, multicentre, phase 3 trial
.
Lancet Oncol
2017
;
18
:
611
22
.
6.
Lebbe
C
,
Meyer
N
,
Mortier
L
,
Marquez-Rodas
I
,
Robert
C
,
Rutkowski
P
, et al
Evaluation of two dosing regimens for nivolumab in combination with ipilimumab in patients with advanced melanoma: results from the phase IIIb/IV CheckMate 511 trial
.
J Clin Oncol
2019
;
37
:
867
75
.
7.
Sharma
P
,
Hu-Lieskovan
S
,
Wargo
JA
,
Ribas
A
. 
Primary, adaptive, and acquired resistance to cancer immunotherapy
.
Cell
2017
;
168
:
707
23
.
8.
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
.
9.
Postow
MA
,
Chesney
J
,
Pavlick
AC
,
Robert
C
,
Grossmann
K
,
McDermott
D
, et al
Nivolumab and ipilimumab versus ipilimumab in untreated melanoma
.
N Engl J Med
2015
;
372
:
2006
17
.
10.
Wolchok
JD
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Rutkowski
P
,
Grob
JJ
,
Cowey
CL
, et al
Overall survival with combined nivolumab and ipilimumab in advanced melanoma
.
N Engl J Med
2017
;
377
:
1345
56
.
11.
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
.
12.
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
.
13.
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
.
14.
Lenz
H-J
,
Lonardi
S
,
Zagonel
V
,
Cutse
EV
,
Limon
ML
,
Wong
KYM
, et al
Nivolumab plus low-dose ipilimumab as first-line therapy in microsatellite instability-high/DNA mismatch repair deficient metastatic colorectal cancer: clinical update
.
J Clin Oncol
2020
;
38
(
suppl 4; abstr 11
).
15.
Overman
MJ
,
McDermott
R
,
Leach
JL
,
Lonardi
S
,
Lenz
HJ
,
Morse
MA
, et al
Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study
.
Lancet Oncol
2017
;
18
:
1182
91
.
16.
Motzer
RJ
,
Tannir
NM
,
McDermott
DF
,
Aren 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
.
17.
Motzer
RJ
,
Rini
BI
,
McDermott
DF
,
Arén Frontera
O
,
Hammers
HJ
,
Carducci
MA
, et al
Nivolumab plus ipilimumab versus sunitinib in first-line treatment for advanced renal cell carcinoma: extended follow-up of efficacy and safety results from a randomised, controlled, phase 3 trial
.
Lancet Oncol
2019
;
20
:
1370
85
.
18.
Lebbé
C
,
Meyer
N
,
Mortier
L
,
Marquez-Rodas
I
,
Robert
C
,
Rutkowski
P
, et al
LBA47: initial results from a phase IIIb/IV study evaluating two dosing regimens of nivolumab (NIVO) in combination with ipilimumab (IPI) in patients with advanced melanoma (CheckMate 511)
.
Ann Oncol
2018
;
29
:
viii737
.
19.
Villanueva
A
. 
Hepatocellular carcinoma
.
N Engl J Med
2019
;
380
:
1450
62
.
20.
He
AR
,
Yau
T
,
Hsu
C
,
Kang
Y-K
,
Kim
T-Y
,
Santoro
A
, et al
Nivolumab (NIVO) + ipilimumab (IPI) combination therapy in patients (pts) with advanced hepatocellular carcinoma (aHCC): subgroup analyses from CheckMate 040
.
J Clin Oncol
2020
;
38
:
512
.
21.
Yau
T
,
Kang
Y-K
,
Kim
T-Y
,
El-Khoueiry
AB
,
Santoro
A
,
Sangro
B
, et al
Nivolumab (NIVO) + ipilimumab (IPI) combination therapy in patients (pts) with advanced hepatocellular carcinoma (aHCC): results from CheckMate 040
.
J Clin Oncol
2019
;
37
:
4012
.
22.
Gandhi
L
,
Rodriguez-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
.
23.
Langer
CJ
,
Gadgeel
SM
,
Borghaei
H
,
Papadimitrakopoulou
VA
,
Patnaik
A
,
Powell
SF
, et al
Carboplatin and pemetrexed with or without pembrolizumab for advanced, non-squamous non-small-cell lung cancer: a randomised, phase 2 cohort of the open-label KEYNOTE-021 study
.
Lancet Oncol
2016
;
17
:
1497
508
.
24.
Gadgeel
S
,
Rodríguez-Abreu
D
,
Speranza
G
,
Esteban
E
,
Felip
E
,
Dómine
M
, et al
Updated analysis from KEYNOTE-189: pembrolizumab or placebo plus pemetrexed and platinum for previously untreated metastatic nonsquamous non-small-cell lung cancer
.
J Clin Oncol
2020
;
38
:
1505
17
.
25.
Paz-Ares
L
,
Luft
A
,
Vicente
D
,
Tafreshi
A
,
Gumus
M
,
Mazieres
J
, et al
Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer
.
N Engl J Med
2018
;
379
:
2040
51
.
26.
Paz-Ares
L
,
Vicente
D
,
Tafreshi
A
,
Robinson
A
,
Parra
HS
,
Mazières
J
, et al
A randomized, placebo-controlled trial of pembrolizumab plus chemotherapy in patients with metastatic squamous non-small-cell lung cancer: protocol-specified final analysis of KEYNOTE-407
.
J Thorac Oncol
2020
;
15
:
1657
69
.
27.
Antonia
SJ
,
Villegas
A
,
Daniel
D
,
Vicente
D
,
Murakami
S
,
Hui
R
, et al
Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC
.
N Engl J Med
2018
;
379
:
2342
50
.
28.
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
.
29.
Gray
JE
,
Villegas
A
,
Daniel
D
,
Vicente
D
,
Murakami
S
,
Hui
R
, et al
Three-year overall survival with durvalumab after chemoradiotherapy in stage III NSCLC-update from PACIFIC
.
J Thorac Oncol
2020
;
15
:
288
93
.
30.
Gadgeel
SM
,
Pennell
NA
,
Fidler
MJ
,
Halmos
B
,
Bonomi
P
,
Stevenson
J
, et al
Phase II study of maintenance pembrolizumab in patients with extensive-stage small cell lung cancer (SCLC)
.
J Thorac Oncol
2018
;
13
:
1393
9
.
31.
Reck
M
,
Luft
A
,
Szczesna
A
,
Havel
L
,
Kim
SW
,
Akerley
W
, et al
Phase III randomized trial of ipilimumab plus etoposide and platinum versus placebo plus etoposide and platinum in extensive-stage small-cell lung cancer
.
J Clin Oncol
2016
;
34
:
3740
8
.
32.
Horn
L
,
Mansfield
AS
,
Szczesna
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
.
33.
Reck
M
,
Liu
SV
,
Mansfield
AS
,
Mok
TSK
,
Scherpereel
A
,
Reinmuth
N
, et al
IMpower133: updated overall survival (OS) analysis of first-line (1L) atezolizumab (atezo) + carboplatin + etoposide in extensive-stage SCLC (ES-SCLC)
.
Ann Oncol
2019
;
30
:
v710
v1
.
34.
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
.
35.
Denkert
C
,
von Minckwitz
G
,
Brase
JC
,
Sinn
BV
,
Gade
S
,
Kronenwett
R
, et al
Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2-positive and triple-negative primary breast cancers
.
J Clin Oncol
2015
;
33
:
983
91
.
36.
Loi
S
,
Michiels
S
,
Salgado
R
,
Sirtaine
N
,
Jose
V
,
Fumagalli
D
, et al
Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial
.
Ann Oncol
2014
;
25
:
1544
50
.
37.
Luo
N
,
Nixon
MJ
,
Gonzalez-Ericsson
PI
,
Sanchez
V
,
Opalenik
SR
,
Li
H
, et al
DNA methyltransferase inhibition upregulates MHC-I to potentiate cytotoxic T lymphocyte responses in breast cancer
.
Nat Commun
2018
;
9
:
248
.
38.
Adams
S
,
Schmid
P
,
Rugo
HS
,
Winer
EP
,
Loirat
D
,
Awada
A
, et al
Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: cohort A of the phase II KEYNOTE-086 study
.
Ann Oncol
2019
;
30
:
397
404
.
39.
Schmid
P
,
Rugo
HS
,
Adams
S
,
Schneeweiss
A
,
Barrios
CH
,
Iwata
H
, et al
Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial
.
Lancet Oncol
2020
;
21
:
44
59
.
40.
Le
X
,
Ferrarotto
R
,
Wise-Draper
T
,
Gillison
M
. 
Evolving role of immunotherapy in recurrent metastatic head and neck cancer
.
J Natl Compr Canc Netw
2020
;
18
:
899
906
.
41.
Chow
LQM
. 
Head and neck cancer
.
N Engl J Med
2020
;
382
:
60
72
.
42.
Burtness
B
,
Harrington
KJ
,
Greil
R
,
Soulières
D
,
Tahara
M
,
de Castro
G
 Jr
, 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
.
43.
Allen
E
,
Jabouille
A
,
Rivera
LB
,
Lodewijckx
I
,
Missiaen
R
,
Steri
V
, et al
Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation
.
Sci Transl Med
2017
;
9
:
eaak9679
.
44.
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
.
45.
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
.
46.
Gainor
JF
,
Shaw
AT
,
Sequist
LV
,
Fu
X
,
Azzoli
CG
,
Piotrowska
Z
, et al
EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer: a retrospective analysis
.
Clin Cancer Res
2016
;
22
:
4585
93
.
47.
Herbst
RS
,
Baas
P
,
Kim
DW
,
Felip
E
,
Perez-Gracia
JL
,
Han
JY
, et al
Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial
.
Lancet
2016
;
387
:
1540
50
.
48.
Rittmeyer
A
,
Barlesi
F
,
Waterkamp
D
,
Park
K
,
Ciardiello
F
,
von Pawel
J
, et al
Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial
.
Lancet
2017
;
389
:
255
65
.
49.
Choueiri
TK
,
Motzer
RJ
. 
Systemic therapy for metastatic renal-cell carcinoma
.
N Engl J Med
2017
;
376
:
354
66
.
50.
Rini
BI
,
Powles
T
,
Atkins
MB
,
Escudier
B
,
McDermott
DF
,
Suarez
C
, et al
Atezolizumab plus bevacizumab versus sunitinib in patients with previously untreated metastatic renal cell carcinoma (IMmotion151): a multicentre, open-label, phase 3, randomised controlled trial
.
Lancet
2019
;
393
:
2404
15
.
51.
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
.
52.
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
.
53.
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
.
54.
Taylor
M
,
Rasco
D
,
Brose
M
. 
A phase 1b/2 trial of lenvatinib plus pembrolizumab in patients with squamous cell carcinoma of the head and neck [abstract]
.
J Clin Oncol
2018
;
36
:
6016
.
55.
Robert
C
,
Grob
JJ
,
Stroyakovskiy
D
,
Karaszewska
B
,
Hauschild
A
,
Levchenko
E
, et al
Five-year outcomes with dabrafenib plus trametinib in metastatic melanoma
.
N Engl J Med
2019
;
381
:
626
36
.
56.
Dummer
R
,
Hauschild
A
,
Santinami
M
,
Atkinson
V
,
Mandalà
M
,
Kirkwood
JM
, et al
Five-year analysis of adjuvant dabrafenib plus trametinib in stage III melanoma
.
N Engl J Med
2020
;
383
:
1139
48
.
57.
Dummer
R
,
Ascierto
PA
,
Gogas
HJ
,
Arance
A
,
Mandala
M
,
Liszkay
G
, et al
Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): a multicentre, open-label, randomised phase 3 trial
.
Lancet Oncol
2018
;
19
:
603
15
.
58.
Nathan
P
,
Dummer
R
,
Long
GV
,
Ascierto
PA
,
Tawbi
HA
,
Robert
C
, et al
LBA43 Spartalizumab plus dabrafenib and trametinib (Sparta-DabTram) in patients (pts) with previously untreated BRAF V600–mutant unresectable or metastatic melanoma: results from the randomized part 3 of the phase III COMBI-i trial
.
Ann Oncol
2020
;
31
:
S1172
.
59.
Melero
I
,
Berman
DM
,
Aznar
MA
,
Korman
AJ
,
Pérez Gracia
JL
,
Haanen
J
. 
Evolving synergistic combinations of targeted immunotherapies to combat cancer
.
Nat Rev Cancer
2015
;
15
:
457
72
.
60.
Ribas
A
,
Puzanov
I
,
Dummer
R
,
Schadendorf
D
,
Hamid
O
,
Robert
C
, et al
Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial
.
Lancet Oncol
2015
;
16
:
908
18
.
61.
De Sousa Linhares
A
,
Battin
C
,
Jutz
S
,
Leitner
J
,
Hafner
C
,
Tobias
J
, et al
Therapeutic PD-L1 antibodies are more effective than PD-1 antibodies in blocking PD-1/PD-L1 signaling
.
Sci Rep
2019
;
9
:
11472
.
62.
Deng
R
,
Bumbaca
D
,
Pastuskovas
CV
,
Boswell
CA
,
West
D
,
Cowan
KJ
, et al
Preclinical pharmacokinetics, pharmacodynamics, tissue distribution, and tumor penetration of anti-PD-L1 monoclonal antibody, an immune checkpoint inhibitor
.
MAbs
2016
;
8
:
593
603
.
63.
Duan
J
,
Cui
L
,
Zhao
X
,
Bai
H
,
Cai
S
,
Wang
G
, et al
Use of immunotherapy with programmed cell death 1 vs programmed cell death ligand 1 inhibitors in patients with cancer: a systematic review and meta-analysis
.
JAMA Oncol
2020
;
6
:
375
84
.
64.
Miyashita
H
,
Mikami
T
,
Satoi
S
,
Cruz
C
,
Galsky
MD
. 
Incidence and risk of colitis with programmed death 1 versus programmed death ligand 1 inhibitors for the treatment of cancer
.
J Immunother
2020
;
43
:
291
8
.
65.
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
.
66.
Duraiswamy
J
,
Kaluza
KM
,
Freeman
GJ
,
Coukos
G
. 
Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors
.
Cancer Res
2013
;
73
:
3591
603
.
67.
Chung
DJ
,
Rossi
M
,
Romano
E
,
Ghith
J
,
Yuan
J
,
Munn
DH
, et al
Indoleamine 2,3-dioxygenase-expressing mature human monocyte-derived dendritic cells expand potent autologous regulatory T cells
.
Blood
2009
;
114
:
555
63
.
68.
Munn
DH
,
Shafizadeh
E
,
Attwood
JT
,
Bondarev
I
,
Pashine
A
,
Mellor
AL
. 
Inhibition of T cell proliferation by macrophage tryptophan catabolism
.
J Exp Med
1999
;
189
:
1363
72
.
69.
Holmgaard
RB
,
Zamarin
D
,
Munn
DH
,
Wolchok
JD
,
Allison
JP
. 
Indoleamine 2,3-dioxygenase is a critical resistance mechanism in antitumor T cell immunotherapy targeting CTLA-4
.
J Exp Med
2013
;
210
:
1389
402
.
70.
Liu
M
,
Wang
X
,
Wang
L
,
Ma
X
,
Gong
Z
,
Zhang
S
, et al
Targeting the IDO1 pathway in cancer: from bench to bedside
.
J Hematol Oncol
2018
;
11
:
100
.
71.
Prendergast
GC
,
Malachowski
WP
,
DuHadaway
JB
,
Muller
AJ
. 
Discovery of IDO1 inhibitors: from bench to bedside
.
Cancer Res
2017
;
77
:
6795
811
.
72.
Labadie
BW
,
Bao
R
,
Luke
JJ
. 
Reimagining IDO pathway inhibition in cancer immunotherapy via downstream focus on the tryptophan-kynurenine-aryl hydrocarbon axis
.
Clin Cancer Res
2019
;
25
:
1462
71
.
73.
Long
GV
,
Dummer
R
,
Hamid
O
,
Gajewski
TF
,
Caglevic
C
,
Dalle
S
, et al
Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study
.
Lancet Oncol
2019
;
20
:
1083
97
.
74.
Blackburn
SD
,
Shin
H
,
Haining
WN
,
Zou
T
,
Workman
CJ
,
Polley
A
, et al
Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection
.
Nat Immunol
2009
;
10
:
29
37
.
75.
Koyama
S
,
Akbay
EA
,
Li
YY
,
Herter-Sprie
GS
,
Buczkowski
KA
,
Richards
WG
, et al
Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints
.
Nat Commun
2016
;
7
:
10501
.
76.
Oweida
A
,
Hararah
MK
,
Phan
A
,
Binder
D
,
Bhatia
S
,
Lennon
S
, et al
Resistance to radiotherapy and PD-L1 blockade is mediated by TIM-3 upregulation and regulatory T-cell infiltration
.
Clin Cancer Res
2018
;
24
:
5368
80
.
77.
Shayan
G
,
Srivastava
R
,
Li
J
,
Schmitt
N
,
Kane
LP
,
Ferris
RL
. 
Adaptive resistance to anti-PD1 therapy by Tim-3 upregulation is mediated by the PI3K-Akt pathway in head and neck cancer
.
Oncoimmunology
2017
;
6
:
e1261779
.
78.
Davar D
BP
,
Eroglu
Z
,
Falchook
G
,
Gainor
J
,
Hamilton
E
,
Hecht
JP
, et al
A phase 1 study of TSR-022, an anti-TIM-3 monoclonal antibody, in combination with TSR-042 (anti-PD-1) in patients with colorectal cancer and post-PD-1 NSCLC and melanoma
.
J Immunother Cancer
2018
;
6
:
O21
.
79.
Harding
JJ
,
Patnaik
A
,
Moreno
V
,
Stein
M
,
Jankowska
AM
,
de Mendizabal
NV
, et al
A phase Ia/Ib study of an anti-TIM-3 antibody (LY3321367) monotherapy or in combination with an anti-PD-L1 antibody (LY3300054): interim safety, efficacy, and pharmacokinetic findings in advanced cancers
.
J Clin Oncol
2019
;
37
:
12
.
80.
Durham
NM
,
Nirschl
CJ
,
Jackson
CM
,
Elias
J
,
Kochel
CM
,
Anders
RA
, et al
Lymphocyte activation gene 3 (LAG-3) modulates the ability of CD4 T-cells to be suppressed in vivo
.
PLoS One
2014
;
9
:
e109080
.
81.
Huang
CT
,
Workman
CJ
,
Flies
D
,
Pan
X
,
Marson
AL
,
Zhou
G
, et al
Role of LAG-3 in regulatory T cells
.
Immunity
2004
;
21
:
503
13
.
82.
Workman
CJ
,
Vignali
DA
. 
Negative regulation of T cell homeostasis by lymphocyte activation gene-3 (CD223)
.
J Immunol
2005
;
174
:
688
95
.
83.
Goding
SR
,
Wilson
KA
,
Xie
Y
,
Harris
KM
,
Baxi
A
,
Akpinarli
A
, et al
Restoring immune function of tumor-specific CD4+ T cells during recurrence of melanoma
.
J Immunol
2013
;
190
:
4899
909
.
84.
Wierz
M
,
Pierson
S
,
Guyonnet
L
,
Viry
E
,
Lequeux
A
,
Oudin
A
, et al
Dual PD1/LAG3 immune checkpoint blockade limits tumor development in a murine model of chronic lymphocytic leukemia
.
Blood
2018
;
131
:
1617
21
.
85.
Woo
SR
,
Turnis
ME
,
Goldberg
MV
,
Bankoti
J
,
Selby
M
,
Nirschl
CJ
, et al
Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape
.
Cancer Res
2012
;
72
:
917
27
.
86.
Hong
DS
,
Schoffski
P
,
Calvo
A
,
Sarantopoulos
J
,
Ochoa De Olza
M
,
Carvajal
RD
, et al
Phase I/II study of LAG525 ± spartalizumab (PDR001) in patients (pts) with advanced malignancies
.
J Clin Oncol
2018
;
36
:
3012
.
87.
Ascierto
PA
,
Bono
P
,
Bhatia
S
,
Melero
I
,
Nyakas
MS
,
Svane
IM
, et al
Efficacy of BMS-986016, a monoclonal antibody that targets lymphocyte activation gene-3 (LAG-3), in combination with nivolumab in pts with melanoma who progressed during prior anti-PD-1/PD-L1 therapy (mel prior IO) in all-comer and biomarker-enriched populations
.
Ann Oncol
2017
;
28
:
v611
v2
.
88.
Lakhani N
BT
,
Abraham
AK
,
Luddy
J
,
Palcza
J
,
Chartash
E
,
Healy
J
, et al
The anti–LAG-3 antibody MK-4280 as monotherapy and in combination with pembrolizumab for advanced solid tumors: first-in-human phase 1 dose-finding study
.
J Immunother cancer
2018
:
O114
.
89.
Weber
J
,
Long
GV
,
Haanen
JB
,
Arance
A
,
Dummer
R
,
Nathan
P
, et al
1304TiP - A randomized, open-label, phase II open platform study evaluating the efficacy and safety of novel spartalizumab (PDR001) combinations in previously treated unresectable or metastatic melanoma (PLATForM)
.
Ann Oncol
2018
;
29
:
viii465
viii6
.
90.
Yu
S
,
Li
A
,
Liu
Q
,
Yuan
X
,
Xu
H
,
Jiao
D
, et al
Recent advances of bispecific antibodies in solid tumors
.
J Hematol Oncol
2017
;
10
:
155
.
91.
Hickingbottom
B
,
Clynes
R
,
Desjarlais
J
,
Li
C
,
Ding
Y
. 
Preliminary safety and pharmacodynamic (PD) activity of XmAb20717, a PD-1 x CTLA-4 bispecific antibody, in a phase I dose escalation study of patients with selected advanced solid tumors
.
J Clin Oncol
2020
;
38
:
e15001
.
92.
Hou
W
,
Yuan
Q
,
Yuan
X
,
Wang
Y
,
Mo
W
,
Wang
H
, et al
A novel tetravalent bispecific antibody targeting programmed death 1 and tyrosine-protein kinase Met for treatment of gastric cancer
.
Invest New Drugs
2019
;
37
:
876
89
.
93.
Li
S
,
Liu
M
,
Do
MH
,
Chou
C
,
Stamatiades
EG
,
Nixon
BG
, et al
Cancer immunotherapy via targeted TGF-β signalling blockade in T(H) cells
.
Nature
2020
;
587
:
121
5
.
94.
Blank
CU
,
Rozeman
EA
,
Fanchi
LF
,
Sikorska
K
,
van de Wiel
B
,
Kvistborg
P
, et al
Neoadjuvant versus adjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma
.
Nat Med
2018
;
24
:
1655
61
.
95.
Chen
G
,
Huang
AC
,
Zhang
W
,
Zhang
G
,
Wu
M
,
Xu
W
, et al
Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response
.
Nature
2018
;
560
:
382
6
.
96.
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
.
97.
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
.
98.
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
.
99.
Schoenfeld
JD
,
Hanna
GJ
,
Jo
VY
,
Rawal
B
,
Chen
YH
,
Catalano
PS
, et al
Neoadjuvant nivolumab or nivolumab plus ipilimumab in untreated oral cavity squamous cell carcinoma: a phase 2 open-label randomized clinical trial
.
JAMA Oncol
2020
;
6
:
1563
70
.
100.
Caudana
P
,
Núñez
NG
,
De La Rochere
P
,
Pinto
A
,
Denizeau
J
,
Alonso
R
, et al
IL2/anti-IL2 complex combined with CTLA-4, but not PD-1, blockade rescues antitumor NK cell function by regulatory T-cell modulation
.
Cancer Immunol Res
2019
;
7
:
443
57
.
101.
Cho
JH
,
Boyman
O
,
Kim
HO
,
Hahm
B
,
Rubinstein
MP
,
Ramsey
C
, et al
An intense form of homeostatic proliferation of naive CD8+ cells driven by IL-2
.
J Exp Med
2007
;
204
:
1787
801
.
102.
Langowski
JL
,
Addepalli
M
,
VanderVeen
L
,
Pena
R
,
Nutakki
R
,
Kirksey
Y
, et al
Abstract B057: the CD122-biased immunostimulatory cytokine NKTR-214 combined with checkpoint blockade leads to mobilization of antitumor immunity and synergistic activity
.
Cancer Immunol Res
2016
;
4
:
B057
.
103.
Diab
A
,
Tannir
NM
,
Bentebibel
SE
,
Hwu
P
,
Papadimitrakopoulou
V
,
Haymaker
C
, et al
Bempegaldesleukin (NKTR-214) plus Nivolumab in Patients with Advanced Solid Tumors: Phase I Dose-Escalation Study of Safety, Efficacy, and Immune Activation (PIVOT-02)
.
Cancer Discov
2020
;
10
:
1158
73
.
104.
Curti
BD
,
Kovacsovics-Bankowski
M
,
Morris
N
,
Walker
E
,
Chisholm
L
,
Floyd
K
, et al
OX40 is a potent immune-stimulating target in late-stage cancer patients
.
Cancer Res
2013
;
73
:
7189
98
.
105.
Goodwin
RG
,
Din
WS
,
Davis-Smith
T
,
Anderson
DM
,
Gimpel
SD
,
Sato
TA
, et al
Molecular cloning of a ligand for the inducible T cell gene 4-1BB: a member of an emerging family of cytokines with homology to tumor necrosis factor
.
Eur J Immunol
1993
;
23
:
2631
41
.
106.
Zappasodi
R
,
Sirard
C
,
Li
Y
,
Budhu
S
,
Abu-Akeel
M
,
Liu
C
, et al
Abstract PR01: mechanistic rationale to combine GITR agonism with PD-1 blockade in cancer patients
.
Cancer Immunol Res
2019
;
7
:
PR01
.
107.
Ribas
A
,
Medina
T
,
Kummar
S
,
Amin
A
,
Kalbasi
A
,
Drabick
JJ
, et al
SD-101 in combination with pembrolizumab in advanced melanoma: results of a phase Ib, multicenter study
.
Cancer Discov
2018
;
8
:
1250
7
.
108.
Reilley
M
,
Tsimberidou
AM
,
Piha-Paul
SA
,
Yap
TA
,
Fu
S
,
Naing
A
, et al
Phase 1 trial of TLR9 agonist lefitolimod in combination with CTLA-4 checkpoint inhibitor ipilimumab in advanced tumors
.
J Clin Oncol
2019
;
37
:
TPS2669
.
109.
Diab
A
,
Haymaker
C
,
Bernatchez
C
,
Andtbacka
RHI
,
Shaheen
M
,
Johnson
D
, et al
1245PD – Intratumoral (IT) Injection of the TLR9 agonist tilsotolimod (IMO-2125) in combination with ipilimumab (ipi) triggers durable responses in PD-1 inhibitor refractory metastatic melanoma (rMM): results from a multicenter, phase I/II study
.
Ann Oncol
2018
;
29
:
viii442
.
110.
Butler
MO
,
Robert
C
,
Negrier
S
,
In
GK
,
Walker
JW
,
Krajsova
I
, et al
ILLUMINATE 301: a randomized phase 3 study of tilsotolimod in combination with ipilimumab compared with ipilimumab alone in patients with advanced melanoma following progression on or after anti-PD-1 therapy
.
J Clin Oncol
2019
;
37
:
TPS9599
.
111.
Meric-Bernstam
F
,
Sandhu
SK
,
Hamid
O
,
Spreafico
A
,
Kasper
S
,
Dummer
R
, et al
Phase Ib study of MIW815 (ADU-S100) in combination with spartalizumab (PDR001) in patients (pts) with advanced/metastatic solid tumors or lymphomas
.
J Clin Oncol
2019
;
37
:
2507
.
112.
Harrington
KJ
,
Brody
J
,
Ingham
M
,
Strauss
J
,
Cemerski
S
,
Wang
M
, et al
LBA15 - Preliminary results of the first-in-human (FIH) study of MK-1454, an agonist of stimulator of interferon genes (STING), as monotherapy or in combination with pembrolizumab (pembro) in patients with advanced solid tumors or lymphomas
.
Ann Oncol
2018
;
29
:
viii712
.
113.
Curran
KJ
,
Pegram
HJ
,
Brentjens
RJ
. 
Chimeric antigen receptors for T cell immunotherapy: current understanding and future directions
.
J Gene Med
2012
;
14
:
405
15
.
114.
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
.
115.
Adusumilli
PS
,
Zauderer
MG
,
Rusch
VW
,
O'Cearbhaill
R
,
Zhu
A
,
Ngai
D
, et al
Regional delivery of mesothelin-targeted CAR T cells for pleural cancers: safety and preliminary efficacy in combination with anti-PD-1 agent
.
J Clin Oncol
2019
;
37
:
2511
.
116.
Rupp
LJ
,
Schumann
K
,
Roybal
KT
,
Gate
RE
,
Ye
CJ
,
Lim
WA
, et al
CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells
.
Sci Rep
2017
;
7
:
737
.
117.
Grosser
R
,
Cherkassky
L
,
Chintala
N
,
Adusumilli
PS
. 
Combination immunotherapy with CAR T cells and checkpoint blockade for the treatment of solid tumors
.
Cancer Cell
2019
;
36
:
471
82
.
118.
Schwartz
HS
,
Grindey
GB
. 
Adriamycin and daunorubicin: a comparison of antitumor activities and tissue uptake in mice following immunosuppression
.
Cancer Res
1973
;
33
:
1837
44
.
119.
Pfirschke
C
,
Engblom
C
,
Rickelt
S
,
Cortez-Retamozo
V
,
Garris
C
,
Pucci
F
, et al
Immunogenic chemotherapy sensitizes tumors to checkpoint blockade therapy
.
Immunity
2016
;
44
:
343
54
.
120.
Heinhuis
KM
,
Ros
W
,
Kok
M
,
Steeghs
N
,
Beijnen
JH
,
Schellens
JHM
. 
Enhancing antitumor response by combining immune checkpoint inhibitors with chemotherapy in solid tumors
.
Ann Oncol
2019
;
30
:
219
35
.
121.
Erdal
E
,
Haider
S
,
Rehwinkel
J
,
Harris
AL
,
McHugh
PJ
. 
A prosurvival DNA damage-induced cytoplasmic interferon response is mediated by end resection factors and is limited by Trex1
.
Genes Dev
2017
;
31
:
353
69
.
122.
Fu
J
,
Kanne
DB
,
Leong
M
,
Glickman
LH
,
McWhirter
SM
,
Lemmens
E
, et al
STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade
.
Sci Transl Med
2015
;
7
:
283ra52
.
123.
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
2016
;
109
:
djw199
.
124.
Kodumudi
KN
,
Weber
A
,
Sarnaik
AA
,
Pilon-Thomas
S
. 
Blockade of myeloid-derived suppressor cells after induction of lymphopenia improves adoptive T cell therapy in a murine model of melanoma
.
J Immunol
2012
;
189
:
5147
54
.
125.
Ma
Y
,
Adjemian
S
,
Mattarollo
SR
,
Yamazaki
T
,
Aymeric
L
,
Yang
H
, et al
Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells
.
Immunity
2013
;
38
:
729
41
.
126.
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
.
127.
Nanda
R
,
Liu
MC
,
Yau
C
,
Asare
S
,
Hylton
N
,
Veer
LVt
, et al
Pembrolizumab plus standard neoadjuvant therapy for high-risk breast cancer (BC): results from I-SPY 2
.
J Clin Oncol
2017
;
35
:
506
.
128.
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
.
129.
Schmid
P
,
Cortes
J
,
Pusztai
L
,
McArthur
H
,
Kümmel
S
,
Bergh
J
, et al
Pembrolizumab for early triple-negative breast cancer
.
N Engl J Med
2020
;
382
:
810
21
.
130.
Deng
L
,
Liang
H
,
Xu
M
,
Yang
X
,
Burnette
B
,
Arina
A
, et al
STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors
.
Immunity
2014
;
41
:
843
52
.
131.
Sato
H
,
Niimi
A
,
Yasuhara
T
,
Permata
TBM
,
Hagiwara
Y
,
Isono
M
, et al
DNA double-strand break repair pathway regulates PD-L1 expression in cancer cells
.
Nat Commun
2017
;
8
:
1751
.
132.
Schumacher
TN
,
Schreiber
RD
. 
Neoantigens in cancer immunotherapy
.
Science
2015
;
348
:
69
74
.
133.
Formenti
SC
,
Rudqvist
NP
,
Golden
E
,
Cooper
B
,
Wennerberg
E
,
Lhuillier
C
, et al
Radiotherapy induces responses of lung cancer to CTLA-4 blockade
.
Nat Med
2018
;
24
:
1845
51
.
134.
Shevtsov
M
,
Sato
H
,
Multhoff
G
,
Shibata
A
. 
Novel approaches to improve the efficacy of immuno-radiotherapy
.
Front Oncol
2019
;
9
:
156
.
135.
Vanpouille-Box
C
,
Alard
A
,
Aryankalayil
MJ
,
Sarfraz
Y
,
Diamond
JM
,
Schneider
RJ
, et al
DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity
.
Nat Commun
2017
;
8
:
15618
.
136.
Ko
EC
,
Raben
D
,
Formenti
SC
. 
The integration of radiotherapy with immunotherapy for the treatment of non-small cell lung cancer
.
Clin Cancer Res
2018
;
24
:
5792
806
.
137.
Komatsu
T
,
Nakamura
K
,
Kawase
A
. 
Abscopal effect of nivolumab in a patient with primary lung cancer
.
J Thorac Oncol
2017
;
12
:
e143
e4
.
138.
Yuan
Z
,
Fromm
A
,
Ahmed
KA
,
Grass
GD
,
Yang
GQ
,
Oliver
DE
, et al
Radiotherapy rescue of a nivolumab-refractory immune response in a patient with PD-L1-negative metastatic squamous cell carcinoma of the lung
.
J Thorac Oncol
2017
;
12
:
e135
e6
.
139.
Gratia
M
,
Rodero
MP
,
Conrad
C
,
Bou Samra
E
,
Maurin
M
,
Rice
GI
, et al
Bloom syndrome protein restrains innate immune sensing of micronuclei by cGAS
.
J Exp Med
2019
;
216
:
1199
213
.
140.
Hartlova
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
.
141.
Chabanon
RM
,
Muirhead
G
,
Krastev
DB
,
Adam
J
,
Morel
D
,
Garrido
M
, et al
PARP inhibition enhances tumor cell-intrinsic immunity in ERCC1-deficient non-small cell lung cancer
.
J Clin Invest
2019
;
129
:
1211
28
.
142.
Ding
L
,
Kim
HJ
,
Wang
Q
,
Kearns
M
,
Jiang
T
,
Ohlson
CE
, et al
PARP inhibition elicits STING-dependent antitumor immunity in Brca1-deficient ovarian cancer
.
Cell Rep
2018
;
25
:
2972
80
.
143.
Sen
T
,
Rodriguez
BL
,
Chen
L
,
Corte
CMD
,
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
.
144.
Shen
J
,
Zhao
W
,
Ju
Z
,
Wang
L
,
Peng
Y
,
Labrie
M
, et al
PARPi triggers the STING-dependent immune response and enhances the therapeutic efficacy of immune checkpoint blockade independent of BRCAness
.
Cancer Res
2019
;
79
:
311
9
.
145.
Ding
L
,
Chen
X
,
Xu
X
,
Qian
Y
,
Liang
G
,
Yao
F
, et al
PARP1 suppresses the transcription of PD-L1 by poly(ADP-Ribosyl)ating STAT3
.
Cancer Immunol Res
2019
;
7
:
136
49
.
146.
Stewart
RA
,
Pilie
PG
,
Yap
TA
. 
Development of PARP and immune-checkpoint inhibitor combinations
.
Cancer Res
2018
;
78
:
6717
25
.
147.
Karzai
F
,
VanderWeele
D
,
Madan
RA
,
Owens
H
,
Cordes
LM
,
Hankin
A
, et al
Activity of durvalumab plus olaparib in metastatic castration-resistant prostate cancer in men with and without DNA damage repair mutations
.
J Immunother Cancer
2018
;
6
:
141
.
148.
Yu
EY
,
Massard
C
,
Retz
M
,
Tafreshi
A
,
Carles Galceran
J
,
Hammerer
P
, et al
Keynote-365 cohort A: pembrolizumab (pembro) plus olaparib in docetaxel-pretreated patients (pts) with metastatic castrate-resistant prostate cancer (mCRPC)
.
J Clin Oncol
2019
;
37
:
145
.
149.
Vinayak
S
,
Tolaney
SM
,
Schwartzberg
LS
,
Mita
MM
,
McCann
GA-L
,
Tan
AR
, et al
TOPACIO/Keynote-162: niraparib + pembrolizumab in patients (pts) with metastatic triple-negative breast cancer (TNBC), a phase 2 trial
.
J Clin Oncol
2018
;
36
:
1011
.
150.
Konstantinopoulos
PA
,
Waggoner
SE
,
Vidal
GA
,
Mita
MM
,
Fleming
GF
,
Holloway
RW
, et al
TOPACIO/Keynote-162 (NCT02657889): a phase 1/2 study of niraparib + pembrolizumab in patients (pts) with advanced triple-negative breast cancer or recurrent ovarian cancer (ROC)—Results from ROC cohort
.
J Clin Oncol
2018
;
36
:
106
.
151.
Yap
TAK
,
Krebs
MG
,
Postel-Vinay
S
,
Bang
YJ
,
El-Khoueiry
A
,
Abida
W
, et al
Phase I modular study of AZD6738, a novel oral, potent and selective ataxia telangiectasia Rad3-related (ATR) inhibitor in combination (combo) with carboplatin, olaparib or durvalumab in patients (pts) with advanced cancers
.
Eur J Cancer
2016
;
69
:
S2
.
152.
Friedman
J
,
Morisada
M
,
Sun
L
,
Moore
EC
,
Padget
M
,
Hodge
JW
, et al
Inhibition of WEE1 kinase and cell cycle checkpoint activation sensitizes head and neck cancers to natural killer cell therapies
.
J Immunother Cancer
2018
;
6
:
59
.
153.
Jin
MH
,
Nam
AR
,
Park
JE
,
Bang
JH
,
Bang
YJ
,
Oh
DY
. 
Therapeutic co-targeting of WEE1 and ATM downregulates PD-L1 expression in pancreatic cancer
.
Cancer Res Treat
2020
;
52
:
149
66
.
154.
Zhang
L
,
Xu
LJ
,
Zhu
J
,
Li
J
,
Xue
BX
,
Gao
J
, et al
ATMJAKPDL1 signaling pathway inhibition decreases EMT and metastasis of androgenindependent prostate cancer
.
Mol Med Rep
2018
;
17
:
7045
54
.
155.
Tsai
AK
,
Khan
AY
,
Worgo
CE
,
Wang
LL
,
Liang
Y
,
Davila
E
. 
A multikinase and DNA-PK inhibitor combination immunomodulates melanomas, suppresses tumor progression, and enhances immunotherapies
.
Cancer Immunol Res
2017
;
5
:
790
803
.
156.
Brea
EJ
,
Oh
CY
,
Manchado
E
,
Budhu
S
,
Gejman
RS
,
Mo
G
, et al
Kinase regulation of human MHC class I molecule expression on cancer cells
.
Cancer Immunol Res
2016
;
4
:
936
47
.
157.
Ribas
A
,
Hodi
FS
,
Callahan
M
,
Konto
C
,
Wolchok
J
. 
Hepatotoxicity with combination of vemurafenib and ipilimumab
.
N Engl J Med
2013
;
368
:
1365
6
.
158.
Minor
DR
,
Puzanov
I
,
Callahan
MK
,
Hug
BA
,
Hoos
A
. 
Severe gastrointestinal toxicity with administration of trametinib in combination with dabrafenib and ipilimumab
.
Pigment Cell Melanoma Res
2015
;
28
:
611
2
.
159.
Ascierto
PA
,
Ferrucci
PF
,
Stephens
R
,
Del Vecchio
M
,
Atkinson
V
,
Schmidt
H
, et al
1244O - KEYNOTE-022 Part 3: phase II randomized study of 1L dabrafenib (D) and trametinib (T) plus pembrolizumab (Pembro) or placebo (PBO) for BRAF-mutant advanced melanoma
.
Ann Oncol
2018
;
29
:
viii442
.
160.
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 BRAFV600E mutation-positive melanoma (IMspire150): primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial
.
Lancet
2020
;
395
:
1835
44
.
161.
Bendell
J
,
Ciardiello
F
,
Tabernero
J
,
Tebbutt
N
,
Eng
C
,
Di Bartolomeo
M
, et al
LBA-004: efficacy and safety results from IMblaze370, a randomised phase III study comparing atezolizumab+cobimetinib and atezolizumab monotherapy vs regorafenib in chemotherapy-refractory metastatic colorectal cancer
.
Ann Oncol
2018
;
29
:
v123
.
162.
Kim
K
,
Skora
AD
,
Li
Z
,
Liu
Q
,
Tam
AJ
,
Blosser
RL
, et al
Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells
.
Proc Natl Acad Sci U S A
2014
;
111
:
11774
9
.
163.
Orillion
A
,
Hashimoto
A
,
Damayanti
N
,
Shen
L
,
Adelaiye-Ogala
R
,
Arisa
S
, et al
Entinostat neutralizes myeloid-derived suppressor cells and enhances the antitumor effect of PD-1 inhibition in murine models of lung and renal cell carcinoma
.
Clin Cancer Res
2017
;
23
:
5187
201
.
164.
Zheng
H
,
Zhao
W
,
Yan
C
,
Watson
CC
,
Massengill
M
,
Xie
M
, et al
HDAC inhibitors enhance T-cell chemokine expression and augment response to PD-1 immunotherapy in lung adenocarcinoma
.
Clin Cancer Res
2016
;
22
:
4119
32
.
165.
Sullivan
RJ
,
Moschos
SJ
,
Johnson
ML
,
Opyrchal
M
,
Ordentlich
P
,
Brouwer
S
, et al
Abstract CT072: efficacy and safety of entinostat (ENT) and pembrolizumab (PEMBRO) in patients with melanoma previously treated with anti-PD1 therapy
.
Cancer Res
2019
;
79
:
CT072
.
166.
Chiappinelli
KB
,
Strissel
PL
,
Desrichard
A
,
Li
H
,
Henke
C
,
Akman
B
, et al
Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses
.
Cell
2015
;
162
:
974
86
.
167.
Ørskov
AD
,
Saini
SK
,
Bjerregaard
A-M
,
Otani
H
,
Holmberg
S
,
Hansen
JW
, et al
Activation of T cells specific to endogenous retroviral peptides: possible association with clinical response to azacitidine in myeloid malignancies
.
Blood
2017
;
130
:
4243
.
168.
Daver
N
,
Garcia-Manero
G
,
Basu
S
,
Boddu
PC
,
Alfayez
M
,
Cortes
JE
, et al
Efficacy, safety, and biomarkers of response to azacitidine and nivolumab in relapsed/refractory acute myeloid leukemia: a nonrandomized, open-label, phase II study
.
Cancer Discov
2019
;
9
:
370
83
.
169.
Lee
JJ
,
Sun
W
,
Bahary
N
,
Ohr
J
,
Rhee
JC
,
Stoller
RG
, et al
Phase 2 study of pembrolizumab in combination with azacitidine in subjects with metastatic colorectal cancer
.
J Clin Oncol
2017
;
35
:
3054
.
170.
Alvarez
E
. 
B16 murine melanoma: historical perspective on the development of a solid tumor model
.
In
:
Teicher
BA
,
editor
.
Tumor models in cancer research
.
Totowa, NJ
:
Humana Press
; 
2011
.
p.
79
95
.
171.
Barth
RJ
 Jr
,
Bock
SN
,
Mule
JJ
,
Rosenberg
SA
. 
Unique murine tumor-associated antigens identified by tumor infiltrating lymphocytes
.
J Immunol
1990
;
144
:
1531
7
.
172.
Kim
KJ
,
Kanellopoulos-Langevin
C
,
Merwin
RM
,
Sachs
DH
,
Asofsky
R
. 
Establishment and characterization of BALB/c lymphoma lines with B cell properties
.
J Immunol
1979
;
122
:
549
54
.
173.
Gajewski
TF
,
Markiewicz
MA
,
Uyttenhove
C
. 
The p815 mastocytoma tumor model
.
Curr Protoc Immunol
2001
;
Chapter 20:Unit 20 4
.
174.
Aslakson
CJ
,
Miller
FR
. 
Selective events in the metastatic process defined by analysis of the sequential dissemination of subpopulations of a mouse mammary tumor
.
Cancer Res
1992
;
52
:
1399
405
.
175.
Sutherland
KD
,
Proost
N
,
Brouns
I
,
Adriaensen
D
,
Song
JY
,
Berns
A
. 
Cell of origin of small cell lung cancer: inactivation of Trp53 and Rb1 in distinct cell types of adult mouse lung
.
Cancer Cell
2011
;
19
:
754
64
.
176.
Roby
KF
,
Taylor
CC
,
Sweetwood
JP
,
Cheng
Y
,
Pace
JL
,
Tawfik
O
, et al
Development of a syngeneic mouse model for events related to ovarian cancer
.
Carcinogenesis
2000
;
21
:
585
91
.
177.
Summerhayes
IC
,
Franks
LM
. 
Effects of donor age on neoplastic transformation of adult mouse bladder epithelium in vitro
.
J Natl Cancer Inst
1979
;
62
:
1017
23
.
178.
Corbett
TH
,
Griswold
DP
 Jr
,
Roberts
BJ
,
Peckham
JC
,
Schabel
FM
 Jr
. 
Tumor induction relationships in development of transplantable cancers of the colon in mice for chemotherapy assays, with a note on carcinogen structure
.
Cancer Res
1975
;
35
:
2434
9
.
179.
Wang
M
,
Bronte
V
,
Chen
PW
,
Gritz
L
,
Panicali
D
,
Rosenberg
SA
, et al
Active immunotherapy of cancer with a nonreplicating recombinant fowlpox virus encoding a model tumor-associated antigen
.
J Immunol
1995
;
154
:
4685
92
.
180.
Castle
JC
,
Kreiter
S
,
Diekmann
J
,
Lower
M
,
van de Roemer
N
,
de Graaf
J
, et al
Exploiting the mutanome for tumor vaccination
.
Cancer Res
2012
;
72
:
1081
91
.
181.
Kreiter
S
,
Vormehr
M
,
van de Roemer
N
,
Diken
M
,
Lower
M
,
Diekmann
J
, et al
Mutant MHC class II epitopes drive therapeutic immune responses to cancer
.
Nature
2015
;
520
:
692
6
.
182.
Sellers
RS
,
Clifford
CB
,
Treuting
PM
,
Brayton
C
. 
Immunological variation between inbred laboratory mouse strains: points to consider in phenotyping genetically immunomodified mice
.
Vet Pathol
2012
;
49
:
32
43
.
183.
Krupke
DM
,
Begley
DA
,
Sundberg
JP
,
Richardson
JE
,
Neuhauser
SB
,
Bult
CJ
. 
The mouse tumor biology database: a comprehensive resource for mouse models of human cancer
.
Cancer Res
2017
;
77
:
e67
70
.
184.
Dankort
D
,
Curley
DP
,
Cartlidge
RA
,
Nelson
B
,
Karnezis
AN
,
Damsky
WE
 Jr
, et al
Braf(V600E) cooperates with Pten loss to induce metastatic melanoma
.
Nat Genet
2009
;
41
:
544
52
.
185.
Hooijkaas
A
,
Gadiot
J
,
Morrow
M
,
Stewart
R
,
Schumacher
T
,
Blank
CU
. 
Selective BRAF inhibition decreases tumor-resident lymphocyte frequencies in a mouse model of human melanoma
.
Oncoimmunology
2012
;
1
:
609
17
.
186.
Kimpfler
S
,
Sevko
A
,
Ring
S
,
Falk
C
,
Osen
W
,
Frank
K
, et al
Skin melanoma development in ret transgenic mice despite the depletion of CD25+Foxp3+ regulatory T cells in lymphoid organs
.
J Immunol
2009
;
183
:
6330
7
.
187.
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
.
188.
Le
AX
,
Bernhard
EJ
,
Holterman
MJ
,
Strub
S
,
Parham
P
,
Lacy
E
, et al
Cytotoxic T cell responses in HLA-A2.1 transgenic mice: recognition of HLA alloantigens and utilization of HLA-A2.1 as a restriction element
.
J Immunol
1989
;
142
:
1366
71
.
189.
Lute
KD
,
May
KF
 Jr
,
Lu
P
,
Zhang
H
,
Kocak
E
,
Mosinger
B
, et al
Human CTLA4 knock-in mice unravel the quantitative link between tumor immunity and autoimmunity induced by anti-CTLA-4 antibodies
.
Blood
2005
;
106
:
3127
33
.
190.
Huang
X
,
Zheng
L
,
Ouyang
W
,
Zhang
M
,
An
AX
,
Zhao
J
, et al
Abstract A207: utilizing human OX40 knock-in mice (HuGEMMTM) to assess antitumor efficacy of OX40-agonistic antibodies
.
Mol Cancer Ther
2018
;
17
:
A207
.
191.
Zheng
L
,
Huang
X
,
Ouyang
W
,
Chen
G
,
An
AX
,
Dong
X
, et al
Abstract A203: generation of human TIM3 knock-in mice for preclinical efficacy assessment of therapeutic antibodies
.
Mol Cancer Ther
2018
;
17
:
A203
.
192.
Jangalwe
S
,
Shultz
LD
,
Mathew
A
,
Brehm
MA
. 
Improved B cell development in humanized NOD-scid IL2Rgamma(null) mice transgenically expressing human stem cell factor, granulocyte-macrophage colony-stimulating factor and interleukin-3
.
Immun Inflamm Dis
2016
;
4
:
427
40
.
193.
Saito
Y
,
Ellegast
JM
,
Rafiei
A
,
Song
Y
,
Kull
D
,
Heikenwalder
M
, et al
Peripheral blood CD34(+) cells efficiently engraft human cytokine knock-in mice
.
Blood
2016
;
128
:
1829
33
.
194.
Rongvaux
A
,
Willinger
T
,
Martinek
J
,
Strowig
T
,
Gearty
SV
,
Teichmann
LL
, et al
Development and function of human innate immune cells in a humanized mouse model
.
Nat Biotechnol
2014
;
32
:
364
72
.
195.
Wang
M
,
Yao
LC
,
Cheng
M
,
Cai
D
,
Martinek
J
,
Pan
CX
, et al
Humanized mice in studying efficacy and mechanisms of PD-1-targeted cancer immunotherapy
.
FASEB J
2018
;
32
:
1537
49
.
196.
Capasso
A
,
Lang
J
,
Pitts
TM
,
Jordan
KR
,
Lieu
CH
,
Davis
SL
, et al
Characterization of immune responses to anti-PD-1 mono and combination immunotherapy in hematopoietic humanized mice implanted with tumor xenografts
.
J Immunother Cancer
2019
;
7
:
37
.
197.
Winograd
R
,
Byrne
KT
,
Evans
RA
,
Odorizzi
PM
,
Meyer
AR
,
Bajor
DL
, et al
Induction of T-cell immunity overcomes complete resistance to PD-1 and CTLA-4 blockade and improves survival in pancreatic carcinoma
.
Cancer Immunol Res
2015
;
3
:
399
411
.
198.
Beatty
GL
,
Chiorean
EG
,
Fishman
MP
,
Saboury
B
,
Teitelbaum
UR
,
Sun
W
, et al
CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans
.
Science
2011
;
331
:
1612
6
.
199.
O'Hara
MH
,
O'Reilly
EM
,
Rosemarie
M
,
Varadhachary
G
,
Wainberg
ZA
,
Ko
A
, et al
Abstract CT004: a phase Ib study of CD40 agonistic monoclonal antibody APX005M together with gemcitabine (Gem) and nab-paclitaxel (NP) with or without nivolumab (Nivo) in untreated metastatic ductal pancreatic adenocarcinoma (PDAC) patients
.
Cancer Res
2019
;
79
:
CT004
.
200.
Desnoyers
LR
,
Vasiljeva
O
,
Richardson
JH
,
Yang
A
,
Menendez
EE
,
Liang
TW
, et al
Tumor-specific activation of an EGFR-targeting probody enhances therapeutic index
.
Sci Transl Med
2013
;
5
:
207ra144
.
201.
Naing
A
,
Thistlethwaite
FC
,
Spira
AI
,
Garcia-Corbacho
J
,
Randhawa
M
,
Eskens
F
, et al
CX-072, a PD-L1 probody therapeutic, as monotherapy in patients with advanced solid tumors: preliminary results of PROCLAIM-CX-072
.
J Clin Oncol
2019
;
37
:
2513
.
202.
Hinner
MJ
,
Aiba
RSB
,
Jaquin
TJ
,
Berger
S
,
Dürr
MC
,
Schlosser
C
, et al
Tumor-localized costimulatory T-cell engagement by the 4-1BB/HER2 bispecific antibody-anticalin fusion PRS-343
.
Clin Cancer Res
2019
;
25
:
5878
89
.
203.
Aznar
MA
,
Tinari
N
,
Rullan
AJ
,
Sanchez-Paulete
AR
,
Rodriguez-Ruiz
ME
,
Melero
I
. 
Intratumoral delivery of immunotherapy-act locally, think globally
.
J Immunol
2017
;
198
:
31
9
.
204.
Sagiv-Barfi
I
,
Czerwinski
DK
,
Levy
S
,
Alam
IS
,
Mayer
AT
,
Gambhir
SS
, et al
Eradication of spontaneous malignancy by local immunotherapy
.
Sci Transl Med
2018
;
10
:
eaan4488
.
205.
Ager
CR
,
Reilley
MJ
,
Nicholas
C
,
Bartkowiak
T
,
Jaiswal
AR
,
Curran
MA
. 
Intratumoral STING activation with T-cell checkpoint modulation generates systemic antitumor immunity
.
Cancer Immunol Res
2017
;
5
:
676
84
.
206.
Leone
RD
,
Emens
LA
. 
Targeting adenosine for cancer immunotherapy
.
J Immunother Cancer
2018
;
6
:
57
.
207.
Park
A
,
Stacy
M
. 
Istradefylline for the treatment of Parkinson's disease
.
Expert Opin Pharmacother
2012
;
13
:
111
4
.
208.
Congreve
M
,
Brown
GA
,
Borodovsky
A
,
Lamb
ML
. 
Targeting adenosine A2A receptor antagonism for treatment of cancer
.
Expert Opin Drug Discov
2018
;
13
:
997
1003
.
209.
Pinter
M
,
Jain
RK
. 
Targeting the renin-angiotensin system to improve cancer treatment: implications for immunotherapy
.
Sci Transl Med
2017
;
9
:
eaan5616
.
210.
Rachidi
S
,
Metelli
A
,
Riesenberg
B
,
Wu
BX
,
Nelson
MH
,
Wallace
C
, et al
Platelets subvert T cell immunity against cancer via GARP-TGFbeta axis
.
Sci Immunol
2017
;
2
:
eaai7911
.
211.
Serafini
P
,
Meckel
K
,
Kelso
M
,
Noonan
K
,
Califano
J
,
Koch
W
, et al
Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function
.
J Exp Med
2006
;
203
:
2691
702
.
212.
Kluger
HM
,
Tawbi
HA
,
Ascierto
ML
,
Bowden
M
,
Callahan
MK
,
Cha
E
, et al
Defining tumor resistance to PD-1 pathway blockade: recommendations from the first meeting of the SITC Immunotherapy Resistance Taskforce
.
J Immunother Cancer
2020
;
8
:
e000398
.
213.
Schvartsman
G
,
Ma
J
,
Bassett
RL
 Jr
,
Haydu
LE
,
Amaria
RN
,
Hwu
P
, et al
Incidence, patterns of progression, and outcomes of preexisting and newly discovered brain metastases during treatment with anti-PD-1 in patients with metastatic melanoma
.
Cancer
2019
;
125
:
4193
202
.
214.
Food and Drug Administration H
. 
Cancer clinical trial eligibility criteria: brain metastases guidance for industry
.
Fed Regist
2020
;
85
:
41992
3
.
215.
Mitchell
TC
,
Hamid
O
,
Smith
DC
,
Bauer
TM
,
Wasser
JS
,
Olszanski
AJ
, et al
Epacadostat plus pembrolizumab in patients with advanced solid tumors: phase I results from a multicenter, open-label phase I/II trial (ECHO-202/KEYNOTE-037)
.
J Clin Oncol
2018
;
36
:
3223
30
.
216.
Gibney
GT
,
Hamid
O
,
Lutzky
J
,
Olszanski
AJ
,
Mitchell
TC
,
Gajewski
TF
, et al
Phase 1/2 study of epacadostat in combination with ipilimumab in patients with unresectable or metastatic melanoma
.
J Immunother Cancer
2019
;
7
:
80
.
217.
Leone
RD
,
Zhao
L
,
Englert
JM
,
Sun
IM
,
Oh
MH
,
Sun
IH
, et al
Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion
.
Science
2019
;
366
:
1013
21
.
218.
Daver
N
,
Boddu
P
,
Garcia-Manero
G
,
Yadav
SS
,
Sharma
P
,
Allison
J
, et al
Hypomethylating agents in combination with immune checkpoint inhibitors in acute myeloid leukemia and myelodysplastic syndromes
.
Leukemia
2018
;
32
:
1094
105
.
219.
Wang
X
,
Waschke
BC
,
Woolaver
RA
,
Chen
Z
,
Zhang
G
,
Piscopio
AD
, et al
Histone deacetylase inhibition sensitizes PD1 blockade-resistant B-cell lymphomas
.
Cancer Immunol Res
2019
;
7
:
1318
31
.
220.
Mao
W
,
Ghasemzadeh
A
,
Freeman
ZT
,
Obradovic
A
,
Chaimowitz
MG
,
Nirschl
TR
, et al
Immunogenicity of prostate cancer is augmented by BET bromodomain inhibition
.
J Immunother Cancer
2019
;
7
:
277
.
221.
Lemery
S
,
Keegan
P
,
Pazdur
R
. 
First FDA approval agnostic of cancer site — when a biomarker defines the indication
.
N Engl J Med
2017
;
377
:
1409
12
.
222.
Marabelle
A
,
Le
DT
,
Ascierto
PA
,
Di Giacomo
AM
,
De Jesus-Acosta
A
,
Delord
JP
, et al
Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study
.
J Clin Oncol
2020
;
38
:
1
10
.
223.
Hierro
C
,
Matos
I
,
Martin-Liberal
J
,
Ochoa de Olza
M
,
Garralda
E
. 
Agnostic-histology approval of new drugs in oncology: are we already there?
Clin Cancer Res
2019
;
25
:
3210
9
.
224.
Gandara
DR
,
Paul
SM
,
Kowanetz
M
,
Schleifman
E
,
Zou
W
,
Li
Y
, et al
Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab
.
Nat Med
2018
;
24
:
1441
8
.
225.
Osipov
A
,
Lim
SJ
,
Popovic
A
,
Azad
NS
,
Laheru
DA
,
Zheng
L
, et al
Tumor mutational burden, toxicity, and response of immune checkpoint inhibitors targeting PD(L)1, CTLA-4, and combination: a meta-regression analysis
.
Clin Cancer Res
2020
;
26
:
4842
51
.
226.
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
.
227.
Routy
B
,
Le Chatelier
E
,
Derosa
L
,
Duong
CPM
,
Alou
MT
,
Daillère
R
, et al
Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors
.
Science
2018
;
359
:
91
7
.
228.
Pitt
JM
,
Vétizou
M
,
Waldschmitt
N
,
Kroemer
G
,
Chamaillard
M
,
Boneca
IG
, et al
Fine-tuning cancer immunotherapy: optimizing the gut microbiome
.
Cancer Res
2016
;
76
:
4602
7
.
229.
Davar
D
,
Vetizou
MA
,
Dzutsev
A
,
Badger
J
,
McCullogh
J
,
Menna
C
, et al
Abstract IA38: manipulating the gut microbiome to improve immunotherapy of melanoma
.
Cancer Immunol Res
2019
;
7
:
IA38
.
230.
Hellmann
MD
,
Paz-Ares
L
,
Bernabe Caro
R
,
Zurawski
B
,
Kim
SW
,
Carcereny Costa
E
, et al
Nivolumab plus ipilimumab in advanced non-small-cell lung cancer
.
N Engl J Med
2019
;
381
:
2020
31
.
231.
Reck
M
,
Ciuleanu
T-E
,
Dols
MC
,
Schenker
M
,
Zurawski
B
,
Menezes
J
, et al
Nivolumab (NIVO) + ipilimumab (IPI) + 2 cycles of platinum-doublet chemotherapy (chemo) vs 4 cycles chemo as first-line (1L) treatment (tx) for stage IV/recurrent non-small cell lung cancer (NSCLC): CheckMate 9LA
.
J Clin Oncol
2020
;
38
:
9501
.
232.
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
.
233.
Reck
M
,
Mok
TSK
,
Nishio
M
,
Jotte
RM
,
Cappuzzo
F
,
Orlandi
F
, et al
Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer (IMpower150): key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open-label phase 3 trial
.
Lancet Respir Med
2019
;
7
:
387
401
.
234.
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 trial
.
Lancet Oncol
2019
;
20
:
924
37
.
235.
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
.
236.
Choueiri
TK
,
Motzer
RJ
,
Rini
BI
,
Haanen
J
,
Campbell
MT
,
Venugopal
B
, et al
Updated efficacy results from the JAVELIN Renal 101 trial: first-line avelumab plus axitinib versus sunitinib in patients with advanced renal cell carcinoma
.
Ann Oncol
2020
;
31
:
1030
9
.
237.
Schmittnaegel
M
,
Rigamonti
N
,
Kadioglu
E
,
Cassara
A
,
Wyser Rmili
C
,
Kiialainen
A
, et al
Dual angiopoietin-2 and VEGFA inhibition elicits antitumor immunity that is enhanced by PD-1 checkpoint blockade
.
Sci Transl Med
2017
;
9
:
eaak9670
.
238.
Voron
T
,
Colussi
O
,
Marcheteau
E
,
Pernot
S
,
Nizard
M
,
Pointet
AL
, et al
VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors
.
J Exp Med
2015
;
212
:
139
48
.
239.
Wu
FT
,
Man
S
,
Xu
P
,
Chow
A
,
Paez-Ribes
M
,
Lee
CR
, et al
Efficacy of cotargeting angiopoietin-2 and the VEGF pathway in the adjuvant postsurgical setting for early breast, colorectal, and renal cancers
.
Cancer Res
2016
;
76
:
6988
7000
.
240.
Grasselly
C
,
Denis
M
,
Bourguignon
A
,
Talhi
N
,
Mathe
D
,
Tourette
A
, et al
The antitumor activity of combinations of cytotoxic chemotherapy and immune checkpoint inhibitors is model-dependent
.
Front Immunol
2018
;
9
:
2100
.
241.
Tallon de Lara
P
,
Cecconi
V
,
Hiltbrunner
S
,
Yagita
H
,
Friess
M
,
Bode
B
, et al
Gemcitabine synergizes with immune checkpoint inhibitors and overcomes resistance in a preclinical model and mesothelioma patients
.
Clin Cancer Res
2018
;
24
:
6345
54
.
242.
Burlion
A
,
Ramos
RN
,
Kc
P
,
Sendeyo
K
,
Corneau
A
,
Menetrier-Caux
C
, et al
A novel combination of chemotherapy and immunotherapy controls tumor growth in mice with a human immune system
.
Oncoimmunology
2019
;
8
:
1596005
.
243.
Wang
L
,
Amoozgar
Z
,
Huang
J
,
Saleh
MH
,
Xing
D
,
Orsulic
S
, et al
Decitabine enhances lymphocyte migration and function and synergizes with CTLA-4 blockade in a murine ovarian cancer model
.
Cancer Immunol Res
2015
;
3
:
1030
41
.
244.
Stone
ML
,
Chiappinelli
KB
,
Li
H
,
Murphy
LM
,
Travers
ME
,
Topper
MJ
, et al
Epigenetic therapy activates type I interferon signaling in murine ovarian cancer to reduce immunosuppression and tumor burden
.
Proc Natl Acad Sci U S A
2017
;
114
:
E10981
E90
.
245.
Peng
D
,
Kryczek
I
,
Nagarsheth
N
,
Zhao
L
,
Wei
S
,
Wang
W
, et al
Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy
.
Nature
2015
;
527
:
249
53
.
246.
Rojas
JJ
,
Sampath
P
,
Hou
W
,
Thorne
SH
. 
Defining effective combinations of immune checkpoint blockade and oncolytic virotherapy
.
Clin Cancer Res
2015
;
21
:
5543
51
.
247.
Saha
D
,
Martuza
RL
,
Rabkin
SD
. 
Macrophage polarization contributes to glioblastoma eradication by combination immunovirotherapy and immune checkpoint blockade
.
Cancer Cell
2017
;
32
:
253
67
.
248.
Bourgeois-Daigneault
M-C
,
Roy
DG
,
Aitken
AS
,
El Sayes
N
,
Martin
NT
,
Varette
O
, et al
Neoadjuvant oncolytic virotherapy before surgery sensitizes triple-negative breast cancer to immune checkpoint therapy
.
Sci Transl Med
2018
;
10
:
eaao1641
.
249.
Speranza
MC
,
Passaro
C
,
Ricklefs
F
,
Kasai
K
,
Klein
SR
,
Nakashima
H
, et al
Preclinical investigation of combined gene-mediated cytotoxic immunotherapy and immune checkpoint blockade in glioblastoma
.
Neuro Oncol
2018
;
20
:
225
35
.
250.
Chon
HJ
,
Lee
WS
,
Yang
H
,
Kong
SJ
,
Lee
NK
,
Moon
ES
, et al
Tumor microenvironment remodeling by intratumoral oncolytic vaccinia virus enhances the efficacy of immune-checkpoint blockade
.
Clin Cancer Res
2019
;
25
:
1612
23
.
251.
Verbrugge
I
,
Hagekyriakou
J
,
Sharp
LL
,
Galli
M
,
West
A
,
McLaughlin
NM
, et al
Radiotherapy increases the permissiveness of established mammary tumors to rejection by immunomodulatory antibodies
.
Cancer Res
2012
;
72
:
3163
74
.
252.
Twyman-Saint Victor
C
,
Rech
AJ
,
Maity
A
,
Rengan
R
,
Pauken
KE
,
Stelekati
E
, et al
Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer
.
Nature
2015
;
520
:
373
.
253.
Kroon
P
,
Gadiot
J
,
Peeters
M
,
Gasparini
A
,
Deken
MA
,
Yagita
H
, et al
Concomitant targeting of programmed death-1 (PD-1) and CD137 improves the efficacy of radiotherapy in a mouse model of human BRAFV600-mutant melanoma
.
Cancer Immunol Immunother
2016
;
65
:
753
63
.
254.
Zhao
J
,
Wen
X
,
Tian
L
,
Li
T
,
Xu
C
,
Wen
X
, et al
Irreversible electroporation reverses resistance to immune checkpoint blockade in pancreatic cancer
.
Nat Commun
2019
;
10
:
899
.
255.
Peng
W
,
Liu
C
,
Xu
C
,
Lou
Y
,
Chen
J
,
Yang
Y
, et al
PD-1 blockade enhances T-cell migration to tumors by elevating IFN-gamma inducible chemokines
.
Cancer Res
2012
;
72
:
5209
18
.
256.
Moon
EK
,
Ranganathan
R
,
Eruslanov
E
,
Kim
S
,
Newick
K
,
O'Brien
S
, et al
Blockade of programmed death 1 augments the ability of human T cells engineered to target NY-ESO-1 to control tumor growth after adoptive transfer
.
Clin Cancer Res
2016
;
22
:
436
47
.
257.
Jayaprakash
P
,
Ai
M
,
Liu
A
,
Budhani
P
,
Bartkowiak
T
,
Sheng
J
, et al
Targeted hypoxia reduction restores T cell infiltration and sensitizes prostate cancer to immunotherapy
.
J Clin Invest
2018
;
128
:
5137
49
.
258.
Goel
S
,
DeCristo
MJ
,
Watt
AC
,
BrinJones
H
,
Sceneay
J
,
Li
BB
, et al
CDK4/6 inhibition triggers anti-tumour immunity
.
Nature
2017
;
548
:
471
5
.
259.
Garton
AJ
,
Seibel
S
,
Lopresti-Morrow
L
,
Crew
L
,
Janson
N
,
Mandiyan
S
, et al
Anti-KIT monoclonal antibody treatment enhances the antitumor activity of immune checkpoint inhibitors by reversing tumor-induced immunosuppression
.
Mol Cancer Ther
2017
;
16
:
671
80
.
260.
Seifert
AM
,
Zeng
S
,
Zhang
JQ
,
Kim
TS
,
Cohen
NA
,
Beckman
MJ
, et al
PD-1/PD-L1 blockade enhances T-cell activity and antitumor efficacy of imatinib in gastrointestinal stromal tumors
.
Clin Cancer Res
2017
;
23
:
454
65
.
261.
De Henau
O
,
Rausch
M
,
Winkler
D
,
Campesato
LF
,
Liu
C
,
Cymerman
DH
, et al
Overcoming resistance to checkpoint blockade therapy by targeting PI3Kγ in myeloid cells
.
Nature
2016
;
539
:
443
.
262.
Kaneda
MM
,
Messer
KS
,
Ralainirina
N
,
Li
H
,
Leem
CJ
,
Gorjestani
S
, et al
PI3Kγ is a molecular switch that controls immune suppression
.
Nature
2016
;
539
:
437
.
263.
Homet Moreno
B
,
Mok
S
,
Comin-Anduix
B
,
Hu-Lieskovan
S
,
Ribas
A
. 
Combined treatment with dabrafenib and trametinib with immune-stimulating antibodies for BRAF mutant melanoma
.
OncoImmunology
2016
;
5
:
e1052212
.
264.
Sai
J
,
Owens
P
,
Novitskiy
SV
,
Hawkins
OE
,
Vilgelm
AE
,
Yang
J
, et al
PI3K inhibition reduces mammary tumor growth and facilitates antitumor immunity and anti-PD1 responses
.
Clin Cancer Res
2017
;
23
:
3371
84
.
265.
Kim
SH
,
Roszik
J
,
Cho
SN
,
Ogata
D
,
Milton
DR
,
Peng
W
, et al
The COX2 effector microsomal PGE2 synthase 1 is a regulator of immunosuppression in cutaneous melanoma
.
Clin Cancer Res
2019
;
25
:
1650
63
.
266.
Choi
H
,
Deng
J
,
Li
S
,
Silk
T
,
Dong
L
,
Brea
EJ
, et al
Pulsatile MEK inhibition improves anti-tumor immunity and T cell function in murine Kras mutant lung cancer
.
Cell Rep
2019
;
27
:
806
19
.
267.
Kinkead
HL
,
Hopkins
A
,
Lutz
E
,
Wu
AA
,
Yarchoan
M
,
Cruz
K
, et al
Combining STING-based neoantigen-targeted vaccine with checkpoint modulators enhances antitumor immunity in murine pancreatic cancer
.
JCI Insight
2018
;
3
:
e122857
.
268.
Zaidi
N
,
Quezada
SA
,
Kuroiwa
JMY
,
Zhang
L
,
Jaffee
EM
,
Steinman
RM
, et al
Anti-CTLA-4 synergizes with dendritic cell-targeted vaccine to promote IL3-dependent CD4(+) effector T cell infiltration into murine pancreatic tumors
.
Ann N Y Acad Sci
2019
;
1445
:
62
73
.