Checkpoint inhibitors are being added to standard-of-care chemotherapy in multiple clinical trials. Success has been reported in non–small and small cell lung carcinomas and urothelial, head and neck, gastric, and esophageal cancers, and promising results are already available in triple-negative breast and pancreatic malignancies. The potential mechanisms of synergy include immunogenic tumor cell death, antiangiogenesis, selective depletion of myeloid immunosuppressive cells, and lymphopenia, which reduces regulatory T cells and makes room for proliferation of effector T cells. However, chemotherapy regimens have not been optimized for such combinations, perhaps explaining some recent clinical trial disappointments. Approaches to make the most of chemoimmunotherapy include neoadjuvant and adjuvant schemes.

Significance: Immunotherapy of cancer based on PD-1/PD-L1 blockade has prompted a revolution in cancer clinical management. Evidence in phase III clinical trials already supports combinations of immunotherapy with standard-of-care chemotherapy for a number of malignant diseases. This review focuses on such evidence and provides an overview of the potential synergistic mechanisms of action and the opportunities to optimize chemoimmunotherapy regimens.

Finding the Achilles' heel of metastatic cancer is a formidable task. The first pharmacologic answer comes from the search for chemicals that would selectively damage replicating cells. Compounds achieving such an effect mainly include DNA-damaging agents, those interfering with the DNA enzymatic replicative machinery, and those impairing nucleotide synthesis or mitotic mechanisms. The development of chemotherapy started in the 1950s with remarkable success for Hodgkin lymphoma (1), testicular cancer (2), and acute lymphoid leukemia (3). Curative effects against such diseases were discovered with the realization that synergistic combinations of several drugs were required, and that efficacy comes at the cost of significant toxicity. Over the years, cytotoxic compounds for cancer therapy have been developed in an empiric quest to attain survival advantage for patients in myriads of disease-by-disease clinical trial efforts.

Unfortunately, for the most prevalent epithelial solid malignancies at metastatic stages, chemotherapy at its best offers only modest prolongation of survival. Indeed, the best effects of chemotherapy are obtained when it is delivered in a complementary fashion to radical surgery, either before the operation (neoadjuvant treatment) or after surgery (adjuvant treatment).

Cellular biology observations indicate that most chemotherapy agents in the clinic ultimately trigger the proapoptotic suicidal machinery of cancer cells. However, resistance mechanisms are most often induced as a result of the expression of multidrug transporters that pump out the chemicals, and genetic and epigenetic adaptations to the selective Darwinian pharmacologic pressure.

The current state of the art is that aggressive and poorly tolerated chemotherapy schemes are instigated in induction and maintenance regimens until radiologic cancer progression involving tumor masses thriving or newly appearing in spite of treatment. Doses and intervals have been chosen under the maximal tolerated dose paradigm. More recently, therapies to attain selective chemotherapy by nano-formulated drugs such as albumin-conjugated taxanes (nab-paclitaxel) or the liposomal formulation of anthracyclines (liposomal doxorubicin), or antibody–drug conjugates (ADC; such as TDM-1) have been developed. In line with this, tumor selectivity of the ADC can be further attained by optimized linkers that release the chemotherapy moiety selectively in the tumor cell context, as in the case of trastuzumab-deruxtecan (4), or by protease activatable precursor forms of the antibodies (probodies) conjugated to chemotherapy agents (5).

Unfortunately, despite its potency, chemotherapy largely lacks a bystander tumor-killing effect, and escape and resistance are most frequently possible for the remaining malignant cells at tolerable doses. Paradoxically, full-dose chemotherapy is known to affect replicating immune system cells. Indeed, chemotherapy at high doses can cause immunosuppression (6), and drugs that were originally devised to treat cancer are currently mainly used for autoimmune disorders or to prevent allograft rejection. Nonetheless, a therapeutic window exists that contradicts the dogmatic interpretation that was taught until recently in medical school classrooms, stating that the mechanisms targeted by chemotherapy were exactly the same as those required for adaptive immune responses, which are dependent on clonal lymphocyte expansions.

Following obscure years of intensive research to harness the power of the cytotoxic immune response against cancer, we are witnessing a revolution in cancer therapeutics exploiting the tumor recognition and destruction capabilities of the immune system. Two approaches have yielded unprecedented success: checkpoint inhibitors (7) and adoptive T-cell therapy with chimeric antigen receptors for B cell–derived malignancies (8). Importantly, the most striking therapeutic effects of checkpoint inhibitors have been observed upon combination with chemotherapy or antiangiogenic drugs, whereas adoptive T-cell therapy requires preconditioning regimens of non-myeloablative chemotherapy or total body irradiation (9, 10). A summary of the main mechanisms at the interface of chemotherapy and immunotherapy is shown graphically in Fig. 1.

Figure 1.

Postulated mechanisms of synergy between chemotherapy and immunotherapy. In the upper part, the pros and cons mechanisms of chemotherapy with regard to the immune response are depicted with emphasis on the fact that the pros must outbalance the cons. In the lower part, the main invoked mechanisms of action are schematically represented with the main supportive literature references. MDSC, myeloid-derived suppressor cell; Treg, regulatory T cell.

Figure 1.

Postulated mechanisms of synergy between chemotherapy and immunotherapy. In the upper part, the pros and cons mechanisms of chemotherapy with regard to the immune response are depicted with emphasis on the fact that the pros must outbalance the cons. In the lower part, the main invoked mechanisms of action are schematically represented with the main supportive literature references. MDSC, myeloid-derived suppressor cell; Treg, regulatory T cell.

Close modal

Tumor Debulking by Chemotherapy

Tumor masses are active producers of immunosuppressive substances that inhibit immune responses systemically (11). Hence, any treatment that would cause tumor tissue debulking (surgery, chemotherapy, or radiotherapy) may have a beneficial impact on the overall antitumor immune response and its efficacy (12). Additionally, the bulk and numbers of tumor cells to be cleared by the immune system are likely to have limitations. Reducing tumor mass leaves fewer tumor cells to be cleared by the immune system, thereby also reducing the chances for immunoescape variants.

Immunogenic Cell Death: Chemotherapy Meets Immunology

Apoptosis was perceived as an immunologically silent form of cell death and turnover. However, the group of G. Kroemer realized that tumor cells apoptotically killed under the influence of certain chemotherapeutic drugs or ionizing radiation vaccinated mice against a subsequent lethal inoculi of viable tumor cells (13). Such ensuing immunity is mainly dependent on CD8 T lymphocytes (13). Incisive research on the mechanisms underlying these effects concluded that the immunogenicity of cell death is a function of premortem reticular stress (14) and release of tissue damaging–denoting substances (alarmins) that alert the immune system. Ultimately, tumor cell debris uptake by professional antigen-presenting dendritic cells and their activation/maturation state seem to determine the immune outcome (15). Several hallmarks of immunogenic cell death have been proposed, including exposure of calreticulin on the outer cell surface; release of ATP, Annexin-1, and HMGB1; autophagy; inflammasome activation; induction of type 1 IFN signaling, and release of mitochondrial formyl peptides (16). Interestingly, in vivo or in vitro measurements of such hallmark mechanisms allow drugs to be ranked as inducers of immunogenic cell death (17, 18). It must be acknowledged that most of the evidence corresponds to imperfect mouse models and to correlative studies in human cell line samples. Ultimately, the contribution of these mechanisms needs to be addressed in patient tissues under treatment. An excellent recent review summarizes specific knowledge on immunogenic cell death and immune mechanisms elicited in mouse models by anticancer agents, listing most conventional chemotherapy drugs (18).

The postulated train of events in immunogenic cell death includes release of antigens from the dying or dead cells that is taken up, processed, and presented by specialized antigen-presenting cells termed conventional type 1 dendritic cells (cDC1). Maturation/activation of such cDC1 cells is induced by factors released by the surrounding stressed cells under the influence of toxics. Such phenomena are collectively termed as cross-priming of tumor antigens (19), but it must be acknowledged that formal experimental proof for chemotherapy-enhanced tumor antigen cross-priming in humans remains to be published.

The most exploitable consequence of these immunogenic cell death phenomena is that chemotherapy may result in mechanisms that unleash immunity to tackle malignant cells that could be intrinsically resistant to the chemotherapeutic agent. Nonetheless, the most immunogenic form of cell death is probably far less immunogenic than the immunogenicity following detection of microbial presence (20). Microbial moieties detected by innate pathogen recognition receptors are the physiologic triggers of immunity. Therefore, further combination with locally given or systemically delivered TLR agonists, STING agonists, or virotherapy could be advisable to make the most of the proimmune effects of chemotherapy (21). Of note, off-target effects of chemotherapy may provide adjuvanticity to raise the immunogenicity of tumor antigens, for instance, the reported TLR4 agonist activity of paclitaxel (22).

Depletion of Myeloid-Derived Suppressor Cells, Cancer-Associated Neutrophils, and Macrophages

Not all cells of the immune system fight cancer. In fact, there are leukocyte populations that depress cytotoxic antitumor immunity. Tumors favor a form of leukocyte hematopoiesis that yields myeloid cells phenotypically similar to immature macrophages and neutrophils that in culture and in vivo dampen antitumor T-cell immunity (23, 24). Such myeloid-derived suppressor cells (MDSC) are known to be sensitive to chemotherapeutic agents in mice and humans, which include platinum compounds and DNA alkylating agents (25, 26). The mechanisms that such immunosuppressive myeloid cells deploy to impair T-cell function are multifarious, encompassing production of growth and proangiogenic factors, T-cell damaging enzymatic functions, and neutrophil extracellular trap formation, among others. For instance, cancer vaccines are more effective under the influence of cyclophosphamide or platinum-based regimens (25, 26), because of attenuation of MDSC. Of note, gemcitabine and 5-FU are also reported to effectively deplete MDSCs (27, 28). Intratumoral macrophages with transcriptomic features conceptualized as M2 are clearly protumoral, whereas those conceptualized as M1 may mediate antitumor function. Chemotherapy drugs such as cyclophosphamide and doxorubicin reportedly favor M1 differentiation (29). Another possible explanation is that by damaging or killing malignant cells, chemotherapeutic drugs downregulate or reduce factors derived from tumor cells that otherwise may favor M2 differentiation, thereby potentially acting through indirect effects.

Tumors promote the formation of tumor-supportive myeloid leukocytes with growth factors (i.e., GM-CSF, G-CSF, M-CSF, HGF, etc.) and attraction with chemokines (i.e., CXCL1, CXCL8, CCL2, etc.; refs. 24, 30). Ironically, the frequently prescribed recombinant G-CSF to treat or prevent chemotherapy-induced neutropenia is likely to be deleterious for the proimmune effects of chemotherapy.

Homeostatic Proliferation of Effector T Cells, and Treg Depletion

T lymphocytes enter the cell cycle as a result of two types of stimuli. On the one hand, cognate antigen recognition spurs rapid clonal expansion, while on the other hand, T-cell numbers are homeostatically maintained by a competition for promitotic growth factors such as IL7 and IL15. Accordingly, lymphopenia results in homeostatic proliferation of antigen-naïve and central memory T cells.

Hence, lymphotoxic chemotherapy might result in a paradoxical reshaping of the T-cell repertoire and the differentiation into tumor-attacking cytotoxic T cells. Of note, the window of doses and dose intervals becomes crucial, because too much chemotherapy would destroy lymphocytes undergoing expansion, but at optimal doses may foster specific immunity. This is best perceived in the context of adoptive T-cell therapy approaches, where chemotherapy drugs (usually cyclophosphamide and fludarabine) make room for homeostatic proliferation of adoptively infused cells (9). This mechanism is considered crucial, and efficacy has been correlated to the circulating levels of homeostatic cytokines and the achieved lymphopenia (9, 31).

Among T cells, a CD4+ subset is specialized at reducing the activation and expansion of effector T lymphocytes. Such a subset differentiates in the thymus and is driven by the transcription factor FOXP3. In cancer, such regulatory T cells (Treg) interfere with tumor immunity in mouse models, and their presence in the tumor microenvironment has been correlated with a poorer prognosis (32). Moreover, FOXP3+ Treg cell differentiation can be induced as a result of local factors in the tumor tissue microenvironment, such as TGFβ (33). Tregs are probably specific to self-antigens conferring a certain level of self-tolerance and are frequently engaged in the cell cycle. Interestingly, some chemotherapies such as cyclophosphamide seem to be selective to some extent at killing these FOXP3+ subpopulations (34). However, clinical studies have questioned whether cyclophosphamide could actually induce relevant levels of Treg depletion (35). Dosing and timing may be critical to the immunomodulatory effects of cyclophosphamide, and the concept of medium-dose intermittent chemotherapy has been suggested as optimal (36). In any case, cyclophosphamide is an imperfect Treg-depleting agent, and more precise and effective ways to target this immunosuppressive population are under clinical investigation.

All considered, predosing with low-dose cyclophosphamide is a common adjuvant procedure in experimental cancer vaccines (37). Treg depletion by chemotherapy is also likely to explain the favorable effects on lymphodepletion in TIL adoptive therapy. It must be remembered that efficient Treg depletion in tumor tissues is a pending issue in cancer immunotherapy. Of note, low-dose regimens of cyclophosphamide such as metronomic or oral schedules might derive their benefit from these immune-mediated mechanisms. Nonetheless, clinical proof for the efficacy of cyclophosphamide as an immunotherapy-potentiating tool remains to be established and is matter of debate.

Immunotherapy with PD-1/PD-L1–blocking checkpoint inhibitors induces striking clinical benefit upon combination with VEGF–VEGFR antagonists for metastatic renal cell carcinoma (RCC; refs. 38, 39) and hepatocellular carcinoma (HCC; ref. 40). Indeed, the multityrosine kinase inhibitor axitinib + pembrolizumab or avelumab and the combination of bevacizumab + atezolizumab are market-approved for first-line treatment of advanced RCC and HCC, respectively. These combined therapeutic mechanisms were originally translated to the clinic for melanoma by S. Hodi's team (41) and are related not only to angiogenesis inhibition, but also to the reshaping of myeloid populations and to VEGF effects on innate lymphocytes (42). Although VEGF–VEGFR-targeted inhibitors cannot be considered as chemotherapy, they illustrate the concept that antiangiogenesis in combination with immunotherapy may result in synergistic effects.

In this regard, proliferating vascular cells can be the prey of a variety of chemotherapeutic agents even at low metronomic doses (43) that thereby cause solid-tumor hypoxia and nutrient deprivation. This is a double-edged sword for cancer immunity. It is intriguing that a relative degree of hypoxia increases the activity of cytotoxic T cells (44) while it may also result in a more hostile myeloid cell environment (44). Furthermore, tissue penetration of T lymphocytes across endothelial barriers could be altered for the better or for the worse by chemotherapy pretreatment (42).

Disruption of the Gut Microbiome Barrier

The importance of the abundance and species composition of the gut microbiome for immunotherapy has been intensively studied and found to exert a significant effect on therapeutic outcomes in response to immunotherapies with potential for therapeutic interventions (45). Chemotherapy agents very actively destroying replicating enterocytes result in disruption of the gut mucosal barrier leading to bacilli and microbial biomolecules that reach otherwise sterile tissues (46). Such barrier disruption is also induced by total body irradiation and may exert important effects on cancer immunity, in part depending on the control of the maturation state of dendritic cells even if they are located distant from the gut (47).

It is worth mentioning that chemotherapy often causes febrile neutropenia as a side effect that is frequently treated with broad-spectrum antibiotics, which severely modify the gut microbiota. When considering the potential effects on concomitant immunotherapy, this could be deleterious, neutral, or favorable with regard to antitumor effects. These phenomena deserve attention in chemoimmunotherapy trials and will most probably be deleterious for patients' outcomes (48).

Recently, attention has focused on the presence of bacteria or their nucleic acids in tumor lesions, particularly in metastasis from digestive cancers (49, 50). It would not be surprising that such levels of microbial colonization could be increased by chemotherapy drugs, thereby exerting anticancer or procancer immune effects.

Deleterious Effects of Chemotherapy on Immunity

High-dose chemotherapy beyond a poorly understood threshold results in overall immune functional depression (51). This is presumably the result of elimination of anticancer effector components or the demise of stem-like T cells able to restore the antigen-recognition repertoire. Such an undesirable effect may come from dose levels and continuous dosing without intervals for immune recovery. Certainly, dose and schedules of chemotherapy have not been chosen for being immune system-friendly.

For instance, in an attempt to deal with these immunosuppressive side effects, strategies of extraction, expansion, preservation, and reinfusion of polyclonal peripheral blood T cells have been clinically tested in patients undergoing chemotherapy to improve overall immunocompetence and tumor immune responsiveness (52). These approaches might be especially suitable for combination with checkpoint inhibitors.

In addition, it must be considered that the approach of combining chemotherapy and immunotherapy also leads to increased toxicity, as observed with most combination therapies. For example, in the lung cancer KEYNOTE-189 trial, despite a similar overall rate of adverse events, the discontinuation rate for all treatments or any treatment component was higher in the chemoimmunotherapy combination arm (53).

Examples of already available phase III evidence supporting the notion of chemoimmunotherapy combinations can be found and have been published or recently released in congresses in multiple oncology conditions [Table 1, including HR and P data evidence for overall survival (OS) or progression-free survival (PFS)]. Moreover, hundreds of clinical trials are ongoing or are in the plan/preparation phases in which immunotherapy agents are combined with chemotherapy regimens again across multiple malignant indications (Supplementary Table). Furthermore, many studies on neoadjuvant settings are ongoing with evidence for enhanced pathologic responses on chemoimmunotherapy that remain to be validated in terms of OS consequences. A general appraisal of these types of combinations is quite favorable, and they will likely expand to numerous diseases and disease stages. However, benefit is not universally found, and there are trials failing to report superiority of chemoimmunotherapy over standard-of-care options.

Table 1.

Chemoimmunotherapy clinical phase III trials with disclosed efficacy results

IndicationStageChemotherapy agentImmunotherapy agentNCTRef.Clinical efficacy (ChtIO/Cht)Statistical evidenceFDA approvalClinical benefit
NSCLC MTS 1L Platinum/Pem Pembrolizumab KEYNOTE-189 (NCT02578680) 53 ORR 47.6 vs. 18.9%; PFS: 8.8 m vs. 4.9 m; OS at 12 months 69.2% vs. 49.4% OS: HR 0.49 (CI 0.38–0.64); P < 0.001 Yes Yes 
NSCLC MTS 1L Platinum-based + Pem Atezolizumab IMpower 132 (NCT02657434) 59 ORR 47% vs. 32%; PFS 7.6 m vs. 5.2 m; OS 18.1 m vs. 13.6 m PFS: HR 0.596 (CI 0.494–0.719); P < 0.0001 No Yes 
NSCLC MTS 1L Histology-based ChT Nivolumab CheckMate 227 (NCT02477826) 57 ORR 48.1 vs. 29.3%; PFS 8.7 vs. 5.8 m; OS 18.8 vs. 15.6 m OS: HR 0.86 (CI 0.69–1.08); P = 0.1859 No No 
NSCLC MTS 1L Histology-based ChT Nivolumab + ipilimumab CheckMate 9LA (NCT03215706) 61 ORR 38 vs. 25%; PFS 6.7 vs. 5 m; OS 15.6 vs. 10.9 m; OS at 1-year 63% vs. 47% OS: HR 0.69 (CI 0.55–0.87); P = 0.0006 Yes Yes 
NSCLC MTS 1L Carb + Pacl + Bev Nivolumab TASUKI-52 (ONO-4538-52) 58 PFS 12.12 vs. 8.11 m; OS not mature PFS: HR 0.56 (CI 0.43–0.71); P = 0.0001 No Yes 
NSCLC MTS 1L Carb + Pacl + Bev Atezolizumab IMpower150 (NCT02366143) 63 ORR 63.5 vs. 48%; PFS 8.3 vs. 8.6 m; OS 19.2 vs. 14.7 m OS: HR 0.78 (CI: 0.64–0.96); P = 0.02 Yes Yes 
NSCLCsq MTS 1L Carb + Nab-Pacl/Pacl Pembrolizumab KEYNOTE-407 (NCT02775435) 56 ORR 57.9 vs. 38.4%; PFS 6.4 vs. 4.8 m; OS 15.9 vs. 11.3 m OS: HR 0.64 (CI 0.49–0.85); P < 0.001 Yes Yes 
NSCLCsq MTS 1L Carb + Pacl/Nab-Pacl Atezolizumab IMpower131 (NCT02367794) 60 ORR 49 vs. 41%; PFS 6.3 m vs. 5.6 m; OS 14.2 m vs. 13.5 m PFS: HR 0.71 (CI 0.60–0.85); P = 0.0001 No No 
SCLC MTS 1L Carb + Etop Atezolizumab IMpower133 (NCT02763579) 65 ORR 60.2 vs. 64.4%; PFS 5.2 vs. 4.3 m; OS 12.3 vs. 10.3 m OS: HR 0.70 (CI 0.54–0.91); P = 0.007 Yes Yes 
SCLC MTS 1L Carb + Etop Durvalumab CASPIAN (NCT03043872) 66 ORR 68 vs. 58%; OS 13 vs. 10.3 m OS: HR 0.73 (CI 0.59–0.91); P = 0.0047 Yes Yes 
SCLC MTS 1L Carb/Cis + Etop Pembrolizumab KEYNOTE-604 (NCT03066778) 67 ORR 70.6 vs. 61.8%; PFS 4.5 vs. 4.3 m; OS 10.8 vs. 9.7 m PFS: HR 0.75 (CI 0.61–0.91); P = 0.0023. OS: HR 0.80 (0.64–0.98); P = 0.0164 No Yes? 
Breast TN MTS 1L Nab-Pacl Atezolizumab IMpassion 130 (NCT02425891) 68, 69, 70 PFS: 7.2 vs. 5.5 m; OS (ITT): 21 vs. 18.7 m; OS (PD-L1>1%): 25.4 vs. 17.9 m PFS: HR 0.80 (CI 0.69–0.92); P = 0.002 OS (ITT): HR 0.87 (CI 0.72–1.02); P = 0.078 OS (PD-L>1%): HR 0.67 (CI 0.53–0.86) Yes, PD-L1 (+) No 
Breast TN MTS 1L Pacl Atezolizumab IMpassion 131 (NCT03125902) 71 ORR 54 vs. 47%; PFS 5.7 vs. 5.6 m; OS 19.2 vs. 22.8 m OS: HR 1.12 (CI 0.76–1.65) PFS: HR 0.82 (CI 0.60–1.12) No No 
Breast TN MTS 1L Nab-Pacl or Pacl or Carb + Gem Pembrolizumab KEYNOTE-355 (NCT02819518) 72 PFS (CPS>10) 9.7 vs. 5.6 m; PFS (CPS>1) 7.6 vs. 5.6 m PFS (CPS>10): HR 0.65 (CI 0.49–0.86) P = 0.00411 Yes (PD-L1 CPS>10) Yes 
Breast TN NeoAd Pacl + Cycloph + Doxor/Epi Pembrolizumab KEYNOTE-522 (NCT03036488) 75 pCR 64.8% vs. 51.2% pCR: Inc. 13.6%; CI 5.4 to 21.8; P < 0.001 No Yes 
Breast TN NeoAd Carb + Nab-Pacl Atezolizumab NeoTRIPaPDL1 (NCT02620280) 76 pCR 43.5% vs. 40.8% Not available No No 
Breast TN early NeoAd Pacl + Cycloph + Doxor Atezolizumab IMpassion031 (NCT03197935) 77 pCR 57.6% vs. 41.1% pCR: Inc. 16.5% (5.9, 27.1); 1-sided P = 0.0044 No Yes 
Bladder MTS 1L Gem + Cis/Carb Avelumab (maintenance) JAVELIN Bladder 100 (NCT02603432) 87 OS 21.4 m vs. 14.3 m OS: HR 0.69 (CI 0.56–0.86); 1-sided P = 0.0005 Yes Yes 
Bladder MTS 1L Gem + Cis/Carb Pembrolizumab KEYNOTE-361 (NCT02853305) 89 ORR 54.7 vs. 44.9%; PFS 8.3 vs. 7.1 m; OS 17 vs. 14.3 m PFS: HR 0.78 (IC 0.65–0.93); P = 0.0033. OS: HR 0.86 (IC 0.72–1.02); P = 0.0407 No No 
HNC MTS 1L Carb/Cis + 5-FU or Carb/Cis + 5-FU + Cetux Pembrolizumab KEYNOTE-048 (NCT02358031) 92 ORR 35 vs. 19%; OS (ITT): 13 vs. 10.7 m. OS (CPS≥1): 13.6 vs. 10.4 m. OS (CPS≥20): 14.7 vs. 11 m OS (ITT): HR 0.77 (CI 0.63–0.93); P = 0.0034. OS (CPS≥1): HR 0.65 (CI 0.53–0.80); P < 0.0001. OS (CPS≥20): 0.60 (CI 0.45–0.82); P = 0.0004 Yes CPS≥1 Yes 
Gastric MTS 1L Cis + 5-FU/Cape Pembrolizumab KEYNOTE-062 (NCT02494583) 101 ORR 52.5% vs. 36.7%; PFS (CPS≥1) 6.9 m vs. 6.4 m; OS (CPS≥1) 12.5 vs. 11.1 m OS (CPS≥10) 12.3 vs. 10.8 m OS (CPS≥1): HR, 0.85 (CI 0.70–1.03); P = 0.05. OS (CPS≥10): HR, 0.85 (CI 0.62–1.17); P = 0.16 No No 
Gastric/Esophageal MTS 1L Cape + Oxali/5-FU + Oxali Nivolumab CheckMate-649 (NCT02872116) 102 PFS (PD-L1 CPS≥5) 7.6 vs. 6.1 m; OS (all) 13.8 vs. 11.6 m OS (all): HR 0.80 (CI 0.68–0.94); P = 0.0002.PFS (CPS≥5): HR 0.68 (CI 0.56–0.81); P < 0.0001 No Yes 
Indication Stage Chemotherapy agent Immunotherapy agent NCT Ref. Clinical efficacy (ChtIO/Cht) Statistical evidence FDA approval Clinical benefit 
Gastric MTS 1L S-1 + Oxali/Cape + Oxali Nivolumab ATTRACTION (ONO-4538–37) 103 ORR 57.5 vs. 47.8%; PFS 10.5 vs. 8.3 m; OS 17.5 vs. 17.2 m PFS: HR 0.68 (CI 0.51–0.90); P = 0.0007 OS: HR 0.90 (CI 0.75–1.08); P = 0.257 No Yes 
Esophageal Adjuvant CRT Nivolumab CheckMate 577 (NCT02743494) 104 DFS 22.4 vs. 11 m DFS: HR 0.69 (CI 0.56–0.86); P = 0.0003 No Yes 
Esophageal MTS 1L Cis + 5-FU Pembrolizumab KEYNOTE-590 (NCT03189719) 105 ORR (all) 45 vs. 29.3%; PFS (all) 6.3 vs. 5.8 m; OS (all) 12.4 vs. 9.8 m PFS (all): HR 0.65 (CI, 0.55–0.76); P < 0.0001. OS (all): HR, 0.73 (CI 0.62–0.86); P < 0.0001 No Yes 
Glioblastoma MTS 1L Temoz + RT Nivolumab CheckMate-548 (NCT02667587) [press release]. Princeton, NJ: BMS; May 9, 2019 PFS not improved, OS not mature Results not published No No 
Ovarian NeoAd/Ad Carb + Pacl + Bev Atezolizumab IMagyn050 (NCT03038100) Ann Oncol. 2020;31:S1161–S1162. PFS (ITT) 19.5 vs. 18.6 m; PFS (PD-L1+) 20.8 vs. 18.5 m; OS not mature PFS (ITT): HR 0.92 (CI 0.79–1.07). PFS (PD-L1+): HR 0.80 (CI 0.65–0.99) No No 
IndicationStageChemotherapy agentImmunotherapy agentNCTRef.Clinical efficacy (ChtIO/Cht)Statistical evidenceFDA approvalClinical benefit
NSCLC MTS 1L Platinum/Pem Pembrolizumab KEYNOTE-189 (NCT02578680) 53 ORR 47.6 vs. 18.9%; PFS: 8.8 m vs. 4.9 m; OS at 12 months 69.2% vs. 49.4% OS: HR 0.49 (CI 0.38–0.64); P < 0.001 Yes Yes 
NSCLC MTS 1L Platinum-based + Pem Atezolizumab IMpower 132 (NCT02657434) 59 ORR 47% vs. 32%; PFS 7.6 m vs. 5.2 m; OS 18.1 m vs. 13.6 m PFS: HR 0.596 (CI 0.494–0.719); P < 0.0001 No Yes 
NSCLC MTS 1L Histology-based ChT Nivolumab CheckMate 227 (NCT02477826) 57 ORR 48.1 vs. 29.3%; PFS 8.7 vs. 5.8 m; OS 18.8 vs. 15.6 m OS: HR 0.86 (CI 0.69–1.08); P = 0.1859 No No 
NSCLC MTS 1L Histology-based ChT Nivolumab + ipilimumab CheckMate 9LA (NCT03215706) 61 ORR 38 vs. 25%; PFS 6.7 vs. 5 m; OS 15.6 vs. 10.9 m; OS at 1-year 63% vs. 47% OS: HR 0.69 (CI 0.55–0.87); P = 0.0006 Yes Yes 
NSCLC MTS 1L Carb + Pacl + Bev Nivolumab TASUKI-52 (ONO-4538-52) 58 PFS 12.12 vs. 8.11 m; OS not mature PFS: HR 0.56 (CI 0.43–0.71); P = 0.0001 No Yes 
NSCLC MTS 1L Carb + Pacl + Bev Atezolizumab IMpower150 (NCT02366143) 63 ORR 63.5 vs. 48%; PFS 8.3 vs. 8.6 m; OS 19.2 vs. 14.7 m OS: HR 0.78 (CI: 0.64–0.96); P = 0.02 Yes Yes 
NSCLCsq MTS 1L Carb + Nab-Pacl/Pacl Pembrolizumab KEYNOTE-407 (NCT02775435) 56 ORR 57.9 vs. 38.4%; PFS 6.4 vs. 4.8 m; OS 15.9 vs. 11.3 m OS: HR 0.64 (CI 0.49–0.85); P < 0.001 Yes Yes 
NSCLCsq MTS 1L Carb + Pacl/Nab-Pacl Atezolizumab IMpower131 (NCT02367794) 60 ORR 49 vs. 41%; PFS 6.3 m vs. 5.6 m; OS 14.2 m vs. 13.5 m PFS: HR 0.71 (CI 0.60–0.85); P = 0.0001 No No 
SCLC MTS 1L Carb + Etop Atezolizumab IMpower133 (NCT02763579) 65 ORR 60.2 vs. 64.4%; PFS 5.2 vs. 4.3 m; OS 12.3 vs. 10.3 m OS: HR 0.70 (CI 0.54–0.91); P = 0.007 Yes Yes 
SCLC MTS 1L Carb + Etop Durvalumab CASPIAN (NCT03043872) 66 ORR 68 vs. 58%; OS 13 vs. 10.3 m OS: HR 0.73 (CI 0.59–0.91); P = 0.0047 Yes Yes 
SCLC MTS 1L Carb/Cis + Etop Pembrolizumab KEYNOTE-604 (NCT03066778) 67 ORR 70.6 vs. 61.8%; PFS 4.5 vs. 4.3 m; OS 10.8 vs. 9.7 m PFS: HR 0.75 (CI 0.61–0.91); P = 0.0023. OS: HR 0.80 (0.64–0.98); P = 0.0164 No Yes? 
Breast TN MTS 1L Nab-Pacl Atezolizumab IMpassion 130 (NCT02425891) 68, 69, 70 PFS: 7.2 vs. 5.5 m; OS (ITT): 21 vs. 18.7 m; OS (PD-L1>1%): 25.4 vs. 17.9 m PFS: HR 0.80 (CI 0.69–0.92); P = 0.002 OS (ITT): HR 0.87 (CI 0.72–1.02); P = 0.078 OS (PD-L>1%): HR 0.67 (CI 0.53–0.86) Yes, PD-L1 (+) No 
Breast TN MTS 1L Pacl Atezolizumab IMpassion 131 (NCT03125902) 71 ORR 54 vs. 47%; PFS 5.7 vs. 5.6 m; OS 19.2 vs. 22.8 m OS: HR 1.12 (CI 0.76–1.65) PFS: HR 0.82 (CI 0.60–1.12) No No 
Breast TN MTS 1L Nab-Pacl or Pacl or Carb + Gem Pembrolizumab KEYNOTE-355 (NCT02819518) 72 PFS (CPS>10) 9.7 vs. 5.6 m; PFS (CPS>1) 7.6 vs. 5.6 m PFS (CPS>10): HR 0.65 (CI 0.49–0.86) P = 0.00411 Yes (PD-L1 CPS>10) Yes 
Breast TN NeoAd Pacl + Cycloph + Doxor/Epi Pembrolizumab KEYNOTE-522 (NCT03036488) 75 pCR 64.8% vs. 51.2% pCR: Inc. 13.6%; CI 5.4 to 21.8; P < 0.001 No Yes 
Breast TN NeoAd Carb + Nab-Pacl Atezolizumab NeoTRIPaPDL1 (NCT02620280) 76 pCR 43.5% vs. 40.8% Not available No No 
Breast TN early NeoAd Pacl + Cycloph + Doxor Atezolizumab IMpassion031 (NCT03197935) 77 pCR 57.6% vs. 41.1% pCR: Inc. 16.5% (5.9, 27.1); 1-sided P = 0.0044 No Yes 
Bladder MTS 1L Gem + Cis/Carb Avelumab (maintenance) JAVELIN Bladder 100 (NCT02603432) 87 OS 21.4 m vs. 14.3 m OS: HR 0.69 (CI 0.56–0.86); 1-sided P = 0.0005 Yes Yes 
Bladder MTS 1L Gem + Cis/Carb Pembrolizumab KEYNOTE-361 (NCT02853305) 89 ORR 54.7 vs. 44.9%; PFS 8.3 vs. 7.1 m; OS 17 vs. 14.3 m PFS: HR 0.78 (IC 0.65–0.93); P = 0.0033. OS: HR 0.86 (IC 0.72–1.02); P = 0.0407 No No 
HNC MTS 1L Carb/Cis + 5-FU or Carb/Cis + 5-FU + Cetux Pembrolizumab KEYNOTE-048 (NCT02358031) 92 ORR 35 vs. 19%; OS (ITT): 13 vs. 10.7 m. OS (CPS≥1): 13.6 vs. 10.4 m. OS (CPS≥20): 14.7 vs. 11 m OS (ITT): HR 0.77 (CI 0.63–0.93); P = 0.0034. OS (CPS≥1): HR 0.65 (CI 0.53–0.80); P < 0.0001. OS (CPS≥20): 0.60 (CI 0.45–0.82); P = 0.0004 Yes CPS≥1 Yes 
Gastric MTS 1L Cis + 5-FU/Cape Pembrolizumab KEYNOTE-062 (NCT02494583) 101 ORR 52.5% vs. 36.7%; PFS (CPS≥1) 6.9 m vs. 6.4 m; OS (CPS≥1) 12.5 vs. 11.1 m OS (CPS≥10) 12.3 vs. 10.8 m OS (CPS≥1): HR, 0.85 (CI 0.70–1.03); P = 0.05. OS (CPS≥10): HR, 0.85 (CI 0.62–1.17); P = 0.16 No No 
Gastric/Esophageal MTS 1L Cape + Oxali/5-FU + Oxali Nivolumab CheckMate-649 (NCT02872116) 102 PFS (PD-L1 CPS≥5) 7.6 vs. 6.1 m; OS (all) 13.8 vs. 11.6 m OS (all): HR 0.80 (CI 0.68–0.94); P = 0.0002.PFS (CPS≥5): HR 0.68 (CI 0.56–0.81); P < 0.0001 No Yes 
Indication Stage Chemotherapy agent Immunotherapy agent NCT Ref. Clinical efficacy (ChtIO/Cht) Statistical evidence FDA approval Clinical benefit 
Gastric MTS 1L S-1 + Oxali/Cape + Oxali Nivolumab ATTRACTION (ONO-4538–37) 103 ORR 57.5 vs. 47.8%; PFS 10.5 vs. 8.3 m; OS 17.5 vs. 17.2 m PFS: HR 0.68 (CI 0.51–0.90); P = 0.0007 OS: HR 0.90 (CI 0.75–1.08); P = 0.257 No Yes 
Esophageal Adjuvant CRT Nivolumab CheckMate 577 (NCT02743494) 104 DFS 22.4 vs. 11 m DFS: HR 0.69 (CI 0.56–0.86); P = 0.0003 No Yes 
Esophageal MTS 1L Cis + 5-FU Pembrolizumab KEYNOTE-590 (NCT03189719) 105 ORR (all) 45 vs. 29.3%; PFS (all) 6.3 vs. 5.8 m; OS (all) 12.4 vs. 9.8 m PFS (all): HR 0.65 (CI, 0.55–0.76); P < 0.0001. OS (all): HR, 0.73 (CI 0.62–0.86); P < 0.0001 No Yes 
Glioblastoma MTS 1L Temoz + RT Nivolumab CheckMate-548 (NCT02667587) [press release]. Princeton, NJ: BMS; May 9, 2019 PFS not improved, OS not mature Results not published No No 
Ovarian NeoAd/Ad Carb + Pacl + Bev Atezolizumab IMagyn050 (NCT03038100) Ann Oncol. 2020;31:S1161–S1162. PFS (ITT) 19.5 vs. 18.6 m; PFS (PD-L1+) 20.8 vs. 18.5 m; OS not mature PFS (ITT): HR 0.92 (CI 0.79–1.07). PFS (PD-L1+): HR 0.80 (CI 0.65–0.99) No No 

NOTE: Each clinical trial is referred to according to text bibliography. Abbreviations: 5-FU, 5-Fluorouracile; Bev, bevacizumab; Cape, capecitabine; Carb, carboplatin; Cetux, cetuximab; ChT, chemotherapy; Cis, cisplatin; CPS, PD-L1 combined positive score; CRT, chemoradiotherapy; Cycloph, cyclophosphamide; Doxor, doxorubicin; DFS, disease-free survival; Epi, epirubicin; Etop, etoposide; Gem, gemcitabine; ITT, intention-to-treat; HNC, head and neck cancer; MTS, metastatic; Nab-Pacl, nab-paclitaxel; NeoAd, neoadjuvant; NeoAd/Ad, neoadjuvant and/or adjuvant; NSCLC, non–small cell lung cancer squamous and non-squamous; NSCLCsq, squamous non–small cell lung cancer; ORR, objective response rate; OS, overall survival; Oxali, oxaliplatin; Pacl, paclitaxel; Pem, pemetrexed; PFS, progression-free survival; pCR, pathologic complete response; RT, radiotherapy; Temoz, temozolamide; TN, triple negative.

For this approach, the accepted standard-of-care chemotherapy regimen has been chosen as such in most cases. We have elected not to present a catalog of immune-related effects of chemotherapy agents as published by Galluzzi and colleagues (18), because in the best of cases it would be incomplete and probably inaccurate. Indeed, the immune-effect profile of chemotherapy drugs commonly used in the clinic needs to be investigated in mouse models and in humans over a broad range of doses.

Chemoimmunotherapy of Lung Cancer

The first and foremost success of chemotherapy being combined with immunotherapy came in the field of non–small cell lung cancer (NSCLC). Following approval of PD-1 and PD-L1 antagonists in second-line metastatic or locally advanced NSCLC, a race for approvals as the first line of treatment with immunotherapy agents began. Single-agent immunotherapy found efficacy in first-line therapy against tumors with high (>50%) PD-L1 expression versus platinum-based chemotherapy doublets (54, 55). In this scenario, an obvious approach was to add immunotherapy to the standard chemotherapy regimens and maintenance with anti–PD-1/PD-L1 immunotherapy as suggested by phase II trials. Amazing unprecedented efficacy in terms of OS as compared with chemotherapy was observed for a pembrolizumab + chemotherapy combination either for adenocarcinoma (HR 0.49; P < 0.001; ref. 53) or for squamous histologies (HR 0.64; P < 0.001; ref. 56). Intriguingly, a parallel trial of chemotherapy with nivolumab failed in its primary endpoint of OS to beat chemotherapy with the chemoimmunotherapy combination in the first-line setting (HR 0.86; P = 0.1859; ref. 57). Nonetheless, results from a randomized phase III clinical trial comparing the addition or not of nivolumab to carboplatin + paclitaxel + bevacizumab for first-line nonsquamous NSCLChave recently disclosed superiority for the nivolumab-containing arm in terms of PFS (HR 0.56; P = 0.0001), while the study is yet immature for OS (58).

The anti–PD-L1 monoclonal antibody atezolizumab combined with carboplatin/cisplatin and pemetrexed for first-line metastatic nonsquamous NSCLC improved PFS compared with chemotherapy alone (HR 0.596; P < 0.0001; ref. 59). Similar benefit in PFS for a chemoimmunotherapy combination with atezolizumab in squamous NSCLC came from another phase III trial (HR 0.71; P = 0.0001), but OS was no longer in the chemoimmunotherapy arm (60). Most recently, ipilimumab + nivolumab combined with chemotherapy surpassed chemotherapy alone as a first-line treatment (HR 0.69; P = 0.0006; ref. 61).

Frequent crossover from chemotherapy to immunotherapy did not prevent improved survival, thus supporting the notion that sequencing both approaches is less beneficial. Importantly, benefit extended to patients with tumors presenting low or no expression of PD-L1. Safety profiles of the combination were tolerable, and >grade 3 adverse events were similar in percentages to chemotherapy + placebo (53, 56). However, as mentioned, the discontinuation rate was higher in the chemoimmunotherapy arm in the Keynote-189 trial (53).

Other PD-1/PD-L1–blocking-based combinations, including those with anti-CTLA4 antibodies and VEGF blockade, have also recently obtained FDA approval for first-line treatment of NSCLC. In this regard, the field has widened with the use of ipilimumab + nivolumab that surpasses chemotherapy, especially in those patients with a high tumor mutational burden (62), and atezolizumab + bevacizumab + chemotherapy, which is better than bevacizumab + chemotherapy (HR 0.78; P = 0.02; ref. 63). As mentioned above, nivolumab in combination with chemotherapy + bevacizumab also rendered benefit over standard chemotherapy + bevacizumab (58).

The most astonishing results of immunotherapy in lung cancer have been observed in the neoadjuvant setting, where nivolumab achieved pathologic complete responses in 45% of cases (64). Several phase III clinical trials are evaluating the added benefit of combining immunotherapy and chemotherapy in the neoadjuvant setting (Supplementary Table).

In small cell lung cancer (SCLC), recent results have also documented in first-line treatment that the addition of PD-L1 blockade with atezolizumab (65) or durvalumab (66) to standard platinum + etoposide yields better results than chemotherapy alone (HR 0.70, P = 0.007; HR 0.73, P = 0.0047, respectively, for atezolizumab or durvalumab). The benefit for pembrolizumab with etoposide + carboplatin over chemotherapy alone has been reported in a phase III trial in terms of PFS (HR 0.75; P = 0.0023), but with a final analysis for OS that did not reach the significance boundary, even if a favorable tendency was observed (HR 0.80; P = 0.0164; ref. 67).

The clinical success of these combinations in the NSCLC and SCLC arenas has not been followed by deep mechanistic insight or the identification of predictive or pharmacodynamic biomarkers. The discovery and validation of such markers is crucial and the focus of ongoing efforts.

Chemoimmunotherapy of Breast Cancer

From the different types of carcinoma of the breast, HER2-negative hormone receptor–negative [triple-negative breast cancer (TNBC)] remains a formidable clinical challenge. In the absence of targeted therapy, the standard of care includes several lines of chemotherapy.

Much enthusiasm and expectation were originally raised by the published results of a clinical trial testing first-line treatment of metastatic TNBC using a combination of nab-paclitaxel + atezolizumab that conferred a better PFS for the combination arm over chemotherapy alone (HR 0.80; P = 0.002), which was higher in the PD-L1–positive tumors (68). However, the final results for OS were negative for the intention-to-treat population (HR 0.87; P = 0.078; refs. 69, 70). The clinical benefit has been described to be almost restricted to patients with PD-L1 > 1% (HR 0.67; refs. 69, 70). Given these results, the field underwent a significant degree of confusion, and the FDA issued an alert regarding the marketing approval. Furthermore, a trial that tested paclitaxel in combination with atezolizumab versus paclitaxel in monotherapy also for first-line TNBC has rendered negative results [HR (OS) 1.12; HR (PFS) 0.82; ref. 71]. In this case, the need for immunosuppressive steroids to permit administration of the weekly taxane might have been involved in the lack of superiority for the chemoimmunotherapy.

A phase III clinical trial comparing pembrolizumab in combination with chemotherapy (paclitaxel, nab-paclitaxel, or carboplatin/gemcitabine) versus chemotherapy alone has shown a significant benefit for the chemoimmunotherapy combination in those patients with combined positive score (CPS) ≥ 10 (HR 0.65; P = 0.00411), leading to FDA approval, but results were not statistically significant for patients with CPS ≤ 1 (72). Of important note, numerous phase I–II clinical trials are exploiting chemoimmunotherapy combinations in patients with metastatic TNBC (Supplementary Table).

In early-stage TNBC following diagnostic biopsy, neoadjuvant chemotherapy regimens have been shown to be highly beneficial to avoid relapse (73). Recently, combination results with pembrolizumab added to the neoadjuvant chemotherapy regimens in two randomized phase II–III trials enhanced the percentage of pathologic complete responses. In this phase II trial (I-SPY2), the pathologic complete response rate in the combination arm was 60% compared with 20% in the chemotherapy-alone arm (74). In the phase III clinical trial, the pathologic complete response rate was 64.8% in the pembrolizumab + chemotherapy arm and 51.2% in the chemotherapy arm (estimated treatment difference 13.6%; P < 0.001; ref. 75) and will be probably conducive to a survival benefit. A randomized study with atezolizumab + chemotherapy in a similar neoadjuvant setting was, however, negative (from 40.8% to 43.5%), even in the PD-L1–positive population (76). Nonetheless, a phase III clinical trial of atezolizumab + chemotherapy in early-stage TNBC has concluded benefit for chemoimmunotherapy, with pathologic complete responses seen in 57.6% versus 41.1% (P = 0.0044; ref. 77). Whether these findings translate into OS and PFS benefit needs longer follow-up.

A phase II randomized clinical trial in the neoadjuvant setting that combines durvalumab versus placebo plus chemotherapy (nab-paclitaxel sequenced to epirubicine + cyclophosphamide) did not show a significant improvement in pCR, although those patients who had received durvalumab alone two weeks before starting chemotherapy had a better and significant pCR advantage versus those who did not receive durvalumab (61.0% vs. 41.4%; OR 2.22; 95% CI 1.06–4.64; P = 0.035; ref. 78). These results stress again the importance of an adequate treatment sequencing of chemo and immunotherapy to achieve the best from such combinations.

At present, the field of TNBC awaits clarification on whether there is a benefit of chemoimmunotherapy versus chemotherapy alone. It could be that the type of chemotherapy, the level of expression of PD-L1, and the vast genetic/transcriptomic heterogeneity of TNBC subtypes matter (79). In this regard, taxanes are considered weak inducers of immunogenic cell death, whereas anthracyclines and alkylating agents are considered to be more proimmunogenic (16). Only further translational research will be able to clarify these points.

Chemoimmunotherapy of Genitourinary Tumors

PD-1/PD-L1 immunotherapy is standard therapy in RCC (80). Prior to the advent of checkpoint blockade, the standard of care for first-line clear cell RCC (ccRCC) treatment was not chemotherapy, but rather antiangiogenic tyrosine kinase inhibitors (TKI). Following the usual paradigm of adding immunotherapy to standard-of-care therapy, first-line treatment of metastatic ccRCC now includes combinations of anti–PD-1 with antiangiogenic agents such as axitinib + pembrolizumab (38), avelumab + axitinib (39), and nivolumab + ipilimumab (81), which in all cases outperformed sunitinib. In this disease, further addition of chemotherapy would be complicated because of its lack of efficacy on its own and the potential for increased toxicity.

In contrast, metastatic urothelial carcinoma has clearly benefited to some extent from platinum-based chemotherapy (82). Pembrolizumab and atezolizumab are approved for second-line treatment of metastatic urothelial carcinoma (83, 84, 85) and as first-line treatment for patients who are ineligible for cisplatin (86). Recently, a phase III clinical trial comparing avelumab as maintenance after an induction phase of chemotherapy versus best supportive care resulted in overwhelmingly better results for the immunotherapy over placebo (HR 0.69; one-sided P = 0.0005), thus leading to FDA approval (87). More recently, exploratory biomarkers in biopsies indicated that those patients with indicators of lymphocyte activity benefited the most with avelumab maintenance, whereas benefit was negatively associated with markers of chronic inflammation (88). By contrast, recently released results from a phase III clinical trial in first line failed to meet the statistical thresholds for the benefit of pembrolizumab combined with chemotherapy as compared with chemotherapy alone, even though there was a clear tendency in favor of chemoimmunotherapy [HR (PFS) 0.78; P = 0.0033; HR (OS) 0.86; P = 0.0407; ref. 89]. This trial (KEYNOTE 361) is very interesting in its three-arm design because it compares chemotherapy alone with chemoimmunotherapy and includes a third immunotherapy-alone arm. However, the comparison with the pembrolizumab single-treatment arm is not available yet.

In castration-resistant metastatic prostate cancer, a beneficial effect of checkpoint inhibitors remains to be seen, although taxane chemotherapy is indicated following hormonal therapy failure. Perhaps subgroups of patients such as those with microsatellite-unstable or BRCA-mutant prostate cancer may benefit from the addition of checkpoint inhibitors to chemotherapy, as is being tested in clinical trials (NCT03834506). As mentioned, one of the potential problems for taxanes is the need for them to be accompanied by steroids, which in pharmacodynamics terms may be at odds with checkpoint inhibitors.

Chemoimmunotherapy of Squamous Head and Neck Cancer

Nivolumab and pembrolizumab are approved for second-line treatment of head and neck cancers (90, 91). Conventional first-line treatment involves the use of chemotherapy and anti-EGFR (cetuximab). Pembrolizumab monotherapy is also approved for first-line treatment of patients whose tumors are CPS > 20 (92). In the same trial (KEYNOTE-048), chemotherapy + pembrolizumab was found to be superior to cetuximab + chemotherapy in the intention-to-treat, in CPS > 1, and in CPS > 20 populations in OS (HR 0.77, 0.65, and 0.60, respectively; P = 0.0034, P < 0.0001, P = 0.0004, respectively), although chemoimmunotherapy did not show superior PFS (92). In this regard, it is worth mentioning that immunotherapy seems to enable more durable objective responses in this setting. The nature of this disease offers many opportunities for neoadjuvant or adjuvant chemoimmunotherapy treatments that are already under intensive investigation in early stages of the disease (Supplementary Table).

Chemoimmunotherapy for Gastrointestinal Tumors

Immunotherapy with checkpoint inhibitors has demonstrated clinical benefit in patients with metastatic microsatellite-unstable colorectal cancer (93), gastric cancer (94), and HCC (95). PD-1/PD-L1 immunotherapy alone minimally affects disease response in patients with pancreatic ductal adenocarcinoma (PDAC; ref. 96); even in microsatellite instability–high (MSI-H) cases the response rate is below 20% (97). Chemotherapy regimens are considered first-line standard of care for colorectal cancer (in combination with cetuximab or bevacizumab), gastric cancer, esophageal cancer, and PDAC.

Colorectal Cancer

In metastatic microsatellite-stable (MSS) colorectal cancer, the use of checkpoint inhibitors thus far offers only anecdotal evidence of clinical activity, despite the fact that T-cell infiltration of primary tumors very much determines prognosis (98). However, recent evidence shows that PD-1 blockade in neoadjuvant regimens prior to removal of resectable primary MSS tumors achieves frequent pathologic responses (99). This disease arena might become fertile for combination regimens including novel immunotherapy agents that are postulated to synergize with chemotherapy regimens. As mentioned, for MSI-H metastatic colorectal cancer, pembrolizumab monotherapy has demonstrated improved PFS over standard of care (chemotherapy plus bevacizumab or cetuximab), supporting recent FDA approval for the indication (100).

Gastric and Esophageal Cancer

Recent data released at the European Society for Medical Oncology (ESMO) Virtual Congress 2020 regarding gastroesophageal junction (GEJ) and gastric cancer provide evidence for chemoimmunotherapy in the first-line setting of these diseases. Publication of such evidence in metastatic or advanced gastric cancer is eagerly awaited because communications in congresses seem to be conflicting to some extent with previous observations. A randomized phase III trial (101) concluded that the addition of chemotherapy to pembrolizumab apparently worsens survival over pembrolizumab or chemotherapy as single treatments (ref. 101; Table 1). Although there may be issues in this trial of patient selection, these results alert us to the fact that chemoimmunotherapy will not be universally beneficial. In line with this, it is possible that agents such as 5-fluorouracile, commonly used in digestive tumor regimens, might be deleterious for anticancer immune responses (51), even more so when used as an add-on rather than in sequential strategies. In striking contrast, recent results from another phase III trial evaluating chemoimmunotherapy combination in first line for advanced cancer of the GEJ/gastric cancer with nivolumab have shown benefit in PFS and OS in the prespecified interim analysis in those patients who received the combination and expressed PD-L1 CPS ≥ 5 [HR (OS) 0.80; P = 0.0002, and HR (PFS CPS ≥ 5) 0.68; P < 0.0001; ref. 102], leading to FDA marketing approval. In the same regard, another study in Asian patients comparing nivolumab + chemotherapy versus chemotherapy for first-line advanced GEJ/gastric cancer showed benefit for chemoimmunotherapy in PFS (HR 0.68; P = 0.0007; ref. 103).

In the localized setting of GEJ/gastric cancer following neoadjuvant chemoradiation and post-resection, adjuvant treatment with nivolumab resulted in a disease-free survival benefit as compared with placebo (HR 0.69; P = 0.003; ref. 104).

In advanced esophageal cancer, more specifically in squamous with PD-L1 CPS > 10, there was a very clear benefit for chemoimmunotherapy over chemotherapy alone (HR 0.57; P < 0.0001), although these positive outcomes favoring chemoimmunotherapy are also expanded to the intention-to-treat population (HR 0.73; P < 0.0001; ref. 105). It would have been very interesting to have an immunotherapy-alone arm in this study.

HCC

In the case of HCC, chemotherapy has never been first-line standard of care, whereas TKIs (sorafenib and lenvatinib) still are. However, chemoembolization is often used for localized disease. In advanced stages, PD-L1 blockade has shown remarkable results in combination with VEGF blockade with bevacizumab over sorafenib (40), changing standard of care. In this disease scenario, considered to be off-limits for chemotherapy, the incorporation of moderate doses of chemotherapy to instigate anticancer immunity is to be explored to further increase efficacy.

PDAC

Metastatic and locally advanced PDAC remains a daunting challenge for clinical oncology. As mentioned, immunotherapy with checkpoint inhibitors has not yielded any valuable results, even in MSI-H cases (96, 97). Chemotherapy of different kinds offers very limited OS benefit (106). In the context of chemoimmunotherapy, a phase II clinical trial combining gemcitabine + nab-paclitaxel with or without durvalumab + tremelimumab has recently disclosed no PFS or OS benefit (107). However, there are reasons for hope because radiologic responses were achieved in 32% of 23 metastatic patients given a regimen encompassing pembrolizumab + 5-FU/leucovorin + Onyvide (liposomal pegylated irinotecan) chemotherapy together with an antagonist of the CXCR4 chemokine receptor (108). Further development of this strategy is warranted and should advance to earlier localized stages of this deadly disease.

Chemoimmunotherapy combinations are changing several paradigms in clinical practice. This approach has been simple and consisted merely of standard-of-care chemotherapy regimens being added to immunotherapy and then being compared with chemotherapy plus placebo. Although this is the most feasible and convenient way to clinically develop this approach, we must remember that the elements in the combination have not been optimized at all. This means that doses, sequence of agents, drugs, and schedules are most likely suboptimal with much potential for improvement (Fig. 2). Knowing that chemotherapy was developed on the basis of a maximal tolerated dosing philosophy, it is likely that at such levels chemotherapy agents are causing some degree of immune cell toxicity. This could be affecting the long-term effects of immunotherapy working against the plateau of the tail in the survival curve, which is so dear to patients and physicians. Whenever possible, an immunotherapy-alone arm should be considered as an additional comparator in clinical trials to really ascertain the benefit of chemoimmunotherapy combinations.

Figure 2.

Road map with the main points of improvement in chemotherapy approaches. Schematic representation of the main criteria and elements to further improve the efficacy of chemoimmunotherapy combinations. TCR, T-cell receptor.

Figure 2.

Road map with the main points of improvement in chemotherapy approaches. Schematic representation of the main criteria and elements to further improve the efficacy of chemoimmunotherapy combinations. TCR, T-cell receptor.

Close modal

To move forward, these issues should be reexamined in preclinical models testing the concept that, at least in certain cases, less chemotherapy in combination with immunotherapy might be conducive to better efficacy. Considering the mechanisms of synergy between chemotherapy and immunotherapy (Fig. 1), sequential approaches might be useful, and these need to be empirically developed in animal models and subsequently in clinical trials. In this regard, a detailed characterization in preclinical models of the profile of immune effects exerted by each chemotherapy agent should be undertaken as has been done for immunogenic cell death (18), but also considering direct or indirect effects on immune system cells.

Unconventional exploratory clinical trial designs can be undertaken, as has been done in the TONIC trial for metastatic TNBC, in which preconditioning chemotherapy and radiotherapy regimens were compared, followed by PD-1 blockade with nivolumab. This trial recruited small randomized cohorts of about 10 to 15 patients. Interestingly, doxorubicin showed better objective response rate (ORR) than cisplatin, radiotherapy, or cyclophosphamide (109). Furthermore, presurgical treatments will allow using pathologic responses and time-to-progression as potential surrogate endpoints to eventually determine patient survival benefit.

This issue of sequencing becomes further complicated by the advent of new immunotherapy agents that will be tested in the arena of the new chemoimmunotherapy standards of care. Nonetheless, concomitancy of treatments may allow chemotherapy to prevent cases of hyperprogression upon exposure to checkpoint inhibitors. This is particularly relevant considering that single-agent immunotherapy has been consistently associated with hyperprogressive disease in certain solid malignancies (110).

The role of surgery remains to be reexamined in the context of chemoimmunotherapy (111), because, contrary to general beliefs, debulking in metastatic stages may be beneficial for immunotherapy as a consequence of removing immunosuppressive factors dependent on tumor mass and reducing the amount of disease to be prey of the immune system.

In our opinion, chemoimmunotherapy regimens should advance to first-line treatment in a number of diseases at metastatic stages. The safety profile of chemoimmunotherapy will probably allow the development of combinations in neoadjuvant settings, in which reductions in chemotherapy intensity might be tested. Importantly, it should not be forgotten that improvements in ORR and PFS do not necessarily lead to longer OS, and ultimately the long-term tail under the Kaplan–Meier curve is the most striking value of immunotherapy, and unfortunately the one that potentially can be curtailed by chemotherapeutics.

It is of note that there are abundant reports from experience outside clinical trials that speak of the unexpected objective responses that do occur upon use of subsequent lines of chemotherapy in patients who had recently progressed to checkpoint inhibitors (68).

In chemoimmunotherapy trials, biomarker studies are to be encouraged, even as substudies in larger randomized studies. In our quest to make the most of chemoimmunotherapy, we need to exploit biomarkers in sequential blood and tumor samples. This should provide suitable pharmacodynamic parameters for optimization of dose, schedule, and drug regimens. For instance, benefit of nab-paclitaxel + atezolizumab in TNBC seemed to depend on PD-L1 expression in the tumor microenvironment (69). However, in NSCLC such a correlation is much weaker, and nonexistent for the combination of chemotherapy with pembrolizumab (53) and the combination of ipilimumab + nivolumab + chemotherapy (61). The room for future treatment individualization of chemoimmunotherapies is therefore huge.

All things considered, chemoimmunotherapy has arrived in the clinical setting and will stay for years to come. However, the field is to be considered in its infancy with regard to understanding mechanisms as well as to optimizing/individualizing treatments. Because of our imperfect knowledge, there will be mixtures of successes and failures in large randomized clinical trials. This overall strategy deserves attention back on the laboratory bench and in translational research-oriented clinical trials, because efficacy could most likely be further improved in a meaningful fashion.

D. Salas-Benito reports being employed at the clinical trial unit of Clínica Universidad de Navarra. J.L. Pérez-Gracia reports grants, personal fees, and non-financial support from Roche, BMS, MSD, and Seattle Genetics, grants and personal fees from Ipsen, grants from Eisai, Incyte, and Janssen outside the submitted work. M. Ponz-Sarvisé reports grants and personal fees from BMS and Roche and personal fees from Incyte outside the submitted work. I. Martínez-Forero reports personal fees from MSD during the conduct of the study. E. Castañón reports personal fees from AstraZeneca, Roche, BMS, and MSD outside the submitted work. M.F. Sanmamed reports grants from Roche outside the submitted work. I. Melero reports grants and personal fees from Roche, Alligator, AstraZeneca, Genmab, and Bristol Myers and personal fees from F-Star, Numab, Pharmamar, Gossamer, and Merck Serono outside the submitted work. No disclosures were reported by the other authors.

This work was supported by Miniserio de ciencia e innovación (SAF2017-83267-C2-1-R), Asociación Española contra el cáncer GCB15152947MELE, and PROCROP European commission HORIZON 2020. We are grateful for healthy discussion with Dr. Jesús San Miguel, Dr. Antonio González, Dr. Ignacio Gil Bazo, and Dr. Carlos de Andrea. Professional English editing by Dr. Paul Miller is also acknowledged.

1.
Cannellos
GP
,
Young
RC
,
DeVita
VT
. 
Combination chemotherapy for advanced Hodgkin's disease in relapse following extensive radiotherapy
.
Clin Pharmacol Ther
1972
;
13
:
750
4
.
2.
Peckham
MJ
,
Barrett
A
,
Liew
KH
,
Horwich
A
,
Robinson
B
,
Dobbs
HJ
, et al
The treatment of metastatic germ-cell testicular tumours with bleomycin, etoposide and cisplatin (BEP)
.
Br J Cancer
1983
;
47
:
613
9
.
3.
Spiers
AS
,
Roberts
PD
,
Marsh
GW
,
Parekh
SJ
,
Franklin
AJ
,
Galton
DA
, et al
Acute lymphoblastic leukaemia: cyclical chemotherapy with three combinations of four drugs (COAP-POMP-CART regimen)
.
Br Med J
1975
;
4
:
614
7
.
4.
Nagai
Y
,
Oitate
M
,
Shiozawa
H
,
Ando
O
. 
Comprehensive preclinical pharmacokinetic evaluations of trastuzumab deruxtecan (DS-8201a), a HER2-targeting antibody-drug conjugate, in cynomolgus monkeys
.
Xenobiotica
2019
;
49
:
1086
96
.
5.
Johnson
ML
,
El-Khoueiry
AB
,
Hafez
N
,
Lakhani
NJ
,
Mamdani
H
,
Rodon Ahnert
J
, et al
CX-2029, a PROBODY drug conjugate targeting CD71 (transferrin receptor): Results from a first-in-human study (PROCLAIM-CX-2029) in patients (Pts) with advanced cancer
.
J Clin Oncol
2020
;
38
:
3502
.
6.
Rasmussen
L
,
Arvin
A
. 
Chemotherapy-induced immunosuppression
.
Environ Health Perspect
1982
;
43
:
21
5
.
7.
Ribas
A
,
Wolchok
JD
. 
Cancer immunotherapy using checkpoint blockade
.
Science
2018
;
359
:
1350
5
.
8.
June
CH
,
O'Connor
RS
,
Kawalekar
OU
,
Ghassemi
S
,
Milone
MC
. 
CAR T cell immunotherapy for human cancer
.
Science
2018
;
359
:
1361
5
.
9.
Rosenberg
SA
,
Restifo
NP
. 
Adoptive cell transfer as personalized immunotherapy for human cancer
.
Science
2015
;
348
:
62
8
.
10.
Goff
SL
,
Dudley
ME
,
Citrin
DE
,
Somerville
RP
,
Wunderlich
JR
,
Danforth
DN
, et al
Randomized, prospective evaluation comparing intensity of lymphodepletion before adoptive transfer of tumor-infiltrating lymphocytes for patients with metastatic melanoma
.
J Clin Oncol
2016
;
34
:
2389
97
.
11.
Rabinovich
GA
,
Gabrilovich
D
,
Sotomayor
EM
. 
Immunosuppressive strategies that are mediated by tumor cells
.
Annu Rev Immunol
2007
;
25
:
267
96
.
12.
Russ
AJ
,
Wentworth
L
,
Xu
K
,
Rakhmilevich
A
,
Seroogy
CM
,
Sondel
PM
, et al
Suppression of T-cell expansion by melanoma is exerted on resting cells
.
Ann Surg Oncol
2011
;
18
:
3848
57
.
13.
Casares
N
,
Pequignot
MO
,
Tesniere
A
,
Ghiringhelli
F
,
Roux
S
,
Chaput
N
, et al
Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death
.
J Exp Med
2005
;
202
:
1691
701
.
14.
Bruno
PM
,
Liu
Y
,
Park
GY
,
Murai
J
,
Koch
CE
,
Eisen
TJ
, et al
A subset of platinum-containing chemotherapeutic agents kills cells by inducing ribosome biogenesis stress
.
Nat Med
2017
;
23
:
461
71
.
15.
Vanmeerbeek
I
,
Sprooten
J
,
De Ruysscher
D
,
Tejpar
S
,
Vandenberghe
P
,
Fucikova
J
, et al
Trial watch: chemotherapy-induced immunogenic cell death in immuno-oncology
.
Oncoimmunology
2020
;
9
:
1703449
.
16.
Galluzzi
L
,
Buqué
A
,
Kepp
O
,
Zitvogel
L
,
Kroemer
G
. 
Immunogenic cell death in cancer and infectious disease
.
Nat Rev Immunol
2017
;
17
:
97
111
.
17.
Menger
L
,
Vacchelli
E
,
Adjemian
S
,
Martins
I
,
Ma
Y
,
Shen
S
, et al
Cardiac glycosides exert anticancer effects by inducing immunogenic cell death
.
Sci Transl Med
2012
;
4
:
143ra99
.
18.
Galluzzi
L
,
Humeau
J
,
Buqué
A
,
Zitvogel
L
,
Kroemer
G
. 
Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors
.
Nat Rev Clin Oncol
2020
;
17
:
725
41
.
19.
Sánchez-Paulete
AR
,
Teijeira
A
,
Cueto
FJ
,
Garasa
S
,
Pérez-Gracia
JL
,
Sánchez-Arráez
A
, et al
Antigen cross-presentation and T-cell cross-priming in cancer immunology and immunotherapy
.
Ann Oncol Off J Eur Soc Med Oncol
2017
;
28
:
xii74
.
20.
Medzhitov
R
,
Janeway
CA
. 
Decoding the patterns of self and nonself by the innate immune system
.
Science
2002
;
296
:
298
300
.
21.
Vanpouille-Box
C
,
Hoffmann
JA
,
Galluzzi
L
. 
Pharmacological modulation of nucleic acid sensors – therapeutic potential and persisting obstacles
.
Nat Rev Drug Discov
2019
;
18
:
845
67
.
22.
Wanderley
CW
,
Colón
DF
,
Luiz
JPM
,
Oliveira
FF
,
Viacava
PR
,
Leite
CA
, et al
Paclitaxel reduces tumor growth by reprogramming tumor-associated macrophages to an M1 profile in a TLR4-dependent manner
.
Cancer Res
2018
;
78
:
5891
900
.
23.
Bronte
V
,
Brandau
S
,
Chen
S-H
,
Colombo
MP
,
Frey
AB
,
Greten
TF
, et al
Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards
.
Nat Commun
2016
;
7
:
12150
.
24.
Gabrilovich
DI
. 
Myeloid-derived suppressor cells
.
Cancer Immunol Res
2017
;
5
:
3
8
.
25.
Welters
MJ
,
van der Sluis
TC
,
van Meir
H
,
Loof
NM
,
van Ham
VJ
,
van Duikeren
S
, et al
Vaccination during myeloid cell depletion by cancer chemotherapy fosters robust T cell responses
.
Sci Transl Med
2016
;
8
:
334ra52
.
26.
Melief
CJM
,
Welters
MJP
,
Vergote
I
,
Kroep
JR
,
Kenter
GG
,
Ottevanger
PB
, et al
Strong vaccine responses during chemotherapy are associated with prolonged cancer survival
.
Sci Transl Med
2020
;
12
:
eaaz8235
.
27.
Suzuki
E
. 
Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity
.
Clin Cancer Res
2005
;
11
:
6713
21
.
28.
Vincent
J
,
Mignot
G
,
Chalmin
F
,
Ladoire
S
,
Bruchard
M
,
Chevriaux
A
, et al
5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity
.
Cancer Res
2010
;
70
:
3052
61
.
29.
Buhtoiarov
IN
,
Sondel
PM
,
Wigginton
JM
,
Buhtoiarova
TN
,
Yanke
EM
,
Mahvi
DA
, et al
Anti-tumour synergy of cytotoxic chemotherapy and anti-CD40 plus CpG-ODN immunotherapy through repolarization of tumour-associated macrophages
.
Immunology
2011
;
132
:
226
39
.
30.
Bonavita
E
,
Galdiero
MR
,
Jaillon
S
,
Mantovani
A
. 
Phagocytes as corrupted policemen in cancer-related inflammation
.
Adv Cancer Res
2015
;
128
:
141
71
.
31.
Gattinoni
L
,
Finkelstein
SE
,
Klebanoff
CA
,
Antony
PA
,
Palmer
DC
,
Spiess
PJ
, et al
Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells
.
J Exp Med
2005
;
202
:
907
12
.
32.
Sakaguchi
S
,
Mikami
N
,
Wing
JB
,
Tanaka
A
,
Ichiyama
K
,
Ohkura
N
. 
Regulatory T cells and human disease
.
Annu Rev Immunol
2020
;
38
:
541
66
.
33.
Chen
W
,
Jin
W
,
Hardegen
N
,
Lei
K-J
,
Li
L
,
Marinos
N
, et al
Conversion of peripheral CD4+CD25-naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3
.
J Exp Med
2003
;
198
:
1875
86
.
34.
Zhao
J
,
Cao
Y
,
Lei
Z
,
Yang
Z
,
Zhang
B
,
Huang
B
. 
Selective depletion of CD4+CD25+Foxp3+ regulatory T cells by low-dose cyclophosphamide is explained by reduced intracellular ATP levels
.
Cancer Res
2010
;
70
:
4850
8
.
35.
Ge
Y
,
Domschke
C
,
Stoiber
N
,
Schott
S
,
Heil
J
,
Rom
J
, et al
Metronomic cyclophosphamide treatment in metastasized breast cancer patients: immunological effects and clinical outcome
.
Cancer Immunol Immunother
2012
;
61
:
353
62
.
36.
Wu
J
,
Waxman
DJ
. 
Immunogenic chemotherapy: dose and schedule dependence and combination with immunotherapy
.
Cancer Lett
2018
;
419
:
210
21
.
37.
Le
DT
,
Jaffee
EM
. 
Regulatory T-cell modulation using cyclophosphamide in vaccine approaches: a current perspective
.
Cancer Res
2012
;
72
:
3439
44
.
38.
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
.
39.
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
.
40.
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
.
41.
Wu
X
,
Li
J
,
Connolly
EM
,
Liao
X
,
Ouyang
J
,
Giobbie-Hurder
A
, et al
Combined anti-VEGF and anti-CTLA-4 therapy elicits humoral immunity to galectin-1 which is associated with favorable clinical outcomes
.
Cancer Immunol Res
2017
;
5
:
446
54
.
42.
Fukumura
D
,
Kloepper
J
,
Amoozgar
Z
,
Duda
DG
,
Jain
RK
. 
Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges
.
Nat Rev Clin Oncol
2018
;
15
:
325
40
.
43.
Hanahan
D
,
Bergers
G
,
Bergsland
E
. 
Less is more, regularly: metronomic dosing of cytotoxic drugs can target tumor angiogenesis in mice
.
J Clin Invest
2000
;
105
:
1045
7
.
44.
Labiano
S
,
Palazon
A
,
Melero
I
. 
Immune response regulation in the tumor microenvironment by hypoxia
.
Semin Oncol
2015
;
42
:
378
86
.
45.
Finlay
BB
,
Goldszmid
R
,
Honda
K
,
Trinchieri
G
,
Wargo
J
,
Zitvogel
L
. 
Can we harness the microbiota to enhance the efficacy of cancer immunotherapy?
Nat Rev Immunol
2020
;20(9):522–8.
46.
Viaud
S
,
Saccheri
F
,
Mignot
G
,
Yamazaki
T
,
Daillère
R
,
Hannani
D
, et al
The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide
.
Science
2013
;
342
:
971
6
.
47.
Viaud
S
,
Daillère
R
,
Boneca
IG
,
Lepage
P
,
Langella
P
,
Chamaillard
M
, et al
Gut microbiome and anticancer immune response: really hot Sh*t!
Cell Death Differ
2015
;
22
:
199
214
.
48.
Elkrief
A
,
Derosa
L
,
Kroemer
G
,
Zitvogel
L
,
Routy
B
. 
The negative impact of antibiotics on outcomes in cancer patients treated with immunotherapy: a new independent prognostic factor?
Ann Oncol Off J Eur Soc Med Oncol
2019
;
30
:
1572
9
.
49.
Geller
LT
,
Barzily-Rokni
M
,
Danino
T
,
Jonas
OH
,
Shental
N
,
Nejman
D
, et al
Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine
.
Science
2017
;
357
:
1156
60
.
50.
Walker
SP
,
Tangney
M
,
Claesson
MJ
. 
Sequence-based characterization of intratumoral bacteria—a guide to best practice
.
Front Oncol
2020
;
10
:
179
.
51.
Kazmers
IS
,
Daddona
PE
,
Dalke
AP
,
Kelley
WN
. 
Effect of immunosuppressive agents on human T and B lymphoblasts
.
Biochem Pharmacol
1983
;
32
:
805
10
.
52.
Rapoport
AP
,
Stadtmauer
EA
,
Aqui
N
,
Badros
A
,
Cotte
J
,
Chrisley
L
, et al
Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer
.
Nat Med
2005
;
11
:
1230
7
.
53.
Gandhi
L
,
Rodríguez-Abreu
D
,
Gadgeel
S
,
Esteban
E
,
Felip
E
,
De Angelis
F
, et al
Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer
.
N Engl J Med
2018
;
378
:
2078
92
.
54.
Reck
M
,
Rodríguez-Abreu
D
,
Robinson
AG
,
Hui
R
,
Csőszi
T
,
Fülöp
A
, et al
Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer
.
N Engl J Med
2016
;
375
:
1823
33
.
55.
Reck
M
,
Rodríguez-Abreu
D
,
Robinson
AG
,
Hui
R
,
Csőszi
T
,
Fülöp
A
, et al
Updated analysis of KEYNOTE-024: pembrolizumab versus platinum-based chemotherapy for advanced non-small-cell lung cancer with PD-L1 tumor proportion score of 50% or greater
.
J Clin Oncol
2019
;
37
:
537
46
.
56.
Paz-Ares
L
,
Luft
A
,
Vicente
D
,
Tafreshi
A
,
Gümüş
M
,
Mazières
J
, et al
Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer
.
N Engl J Med
2018
;
379
:
2040
51
.
57.
Paz-Ares
L
,
Ciuleanu
TE
,
Yu
X
,
Salman
P
,
Pluzanski
A
,
Nagrial
A
, et al
LBA3 nivolumab (NIVO) + platinum-doublet chemotherapy (chemo) vs chemo as first-line (1L) treatment (tx) for advanced non-small cell lung cancer (aNSCLC): CheckMate 227 – part 2 final analysis
.
Ann Oncol
2019
;
30
:
xi67
8
.
58.
Lee
J-S
,
Sugawara
S
,
Kang
JH
,
Kim
HR
,
Inui
N
,
Hida
T
, et al
LBA54 Randomized phase III trial of nivolumab in combination with carboplatin, paclitaxel, and bevacizumab as first-line treatment for patients with advanced or recurrent non-squamous NSCLC
.
Ann Oncol
2020
;
31
:
S1184
5
.
59.
Papadimitrakopoulou
V
,
Cobo
M
,
Bordoni
R
,
Dubray-Longeras
P
,
Szalai
Z
,
Ursol
G
, et al
OA05.07 IMpower132: PFS and safety results with 1L atezolizumab + carboplatin/cisplatin + pemetrexed in stage IV non-squamous NSCLC
.
J Thorac Oncol
2018
;
13
:
S332
3
.
60.
Jotte
R
,
Cappuzzo
F
,
Vynnychenko
I
,
Stroyakovskiy
D
,
Rodríguez-Abreu
D
,
Hussein
M
, et al
Atezolizumab in combination with carboplatin and nab-paclitaxel in advanced squamous NSCLC (IMpower131): results from a randomized phase III trial
.
J Thorac Oncol
2020
;
15
:
1351
60
.
61.
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
.
62.
Hellmann
MD
,
Ciuleanu
TE
,
Pluzanski
A
,
Lee
JS
,
Otterson
GA
,
Audigier-Valette
C
, et al
Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden
.
N Engl J Med
2018
;
378
:
2093
104
.
63.
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
.
64.
Forde
PM
,
Chaft
JE
,
Smith
KN
,
Anagnostou
V
,
Cottrell
TR
,
Hellmann
MD
, et al
Neoadjuvant PD-1 blockade in resectable lung cancer
.
N Engl J Med
2018
;
378
:
1976
86
.
65.
Horn
L
,
Mansfield
AS
,
Szczęsna
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
.
66.
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
.
67.
Rudin
CM
,
Awad
MM
,
Navarro
A
,
Gottfried
M
,
Peters
S
,
Csőszi
T
, et al
Pembrolizumab or placebo plus etoposide and platinum as first-line therapy for extensive-stage small-cell lung cancer: randomized, double-blind, phase III KEYNOTE-604 study
.
J Clin Oncol
2020
;
38
:
2369
79
.
68.
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
.
69.
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
.
70.
Emens
LA
,
Adams
S
,
Barrios
CH
,
Dieras
VC
,
Iwata
H
,
Loi
S
, et al
LBA16 IMpassion130: Final OS analysis from the pivotal phase III study of atezolizumab + nab-paclitaxel vs placebo + nab-paclitaxel in previously untreated locally advanced or metastatic triple-negative breast cancer
.
Ann Oncol
2020
;
31
:
S1148
.
71.
Miles
DW
,
Gligorov
J
,
André
F
,
Cameron
D
,
Schneeweiss
A
,
Barrios
CH
, et al
LBA15 Primary results from IMpassion131, a double-blind placebo-controlled randomised phase III trial of first-line paclitaxel (PAC) ± atezolizumab (atezo) for unresectable locally advanced/metastatic triple-negative breast cancer (mTNBC)
.
Ann Oncol
2020
;
31
:
S1147
8
.
72.
Cortes
J
,
Cescon
DW
,
Rugo
HS
,
Nowecki
Z
,
Im
S-A
,
Yusof
MM
, et al
KEYNOTE-355: Randomized, double-blind, phase III study of pembrolizumab + chemotherapy versus placebo + chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer
.
J Clin Oncol
2020
;
38
:
1000
.
73.
Cortazar
P
,
Zhang
L
,
Untch
M
,
Mehta
K
,
Costantino
JP
,
Wolmark
N
, et al
Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis
.
Lancet
2014
;
384
:
164
72
.
74.
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
684
.
75.
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
.
76.
Gianni
L
,
Huang
C-S
,
Egle
D
,
Bermejo
B
,
Zamagni
C
,
Thill
M
, et al
Abstract GS3-04: Pathologic complete response (pCR) to neoadjuvant treatment with or without atezolizumab in triple negative, early high-risk and locally advanced breast cancer. NeoTRIPaPDL1 Michelangelo randomized study
.
Gen Sess Abstr AACR
; 
2020
:
GS3-04
GS3-04
.
77.
Harbeck
N
,
Zhang
H
,
Barrios
CH
,
Saji
S
,
Jung
KH
,
Hegg
R
, et al
LBA11 IMpassion031: Results from a phase III study of neoadjuvant (neoadj) atezolizumab + chemotherapy in early triple-negative breast cancer (TNBC)
.
Ann Oncol
2020
;
31
:
S1144
.
78.
Loibl
S
,
Untch
M
,
Burchardi
N
,
Huober
J
,
Sinn
BV
,
Blohmer
J-U
, et al
A randomised phase II study investigating durvalumab in addition to an anthracycline taxane-based neoadjuvant therapy in early triple-negative breast cancer: clinical results and biomarker analysis of GeparNuevo study
.
Ann Oncol
2019
;
30
:
1279
88
.
79.
Baird
RD
,
Caldas
C
. 
Genetic heterogeneity in breast cancer: the road to personalized medicine?
BMC Med
2013
;
11
:
151
.
80.
Motzer
RJ
,
Escudier
B
,
McDermott
DF
,
George
S
,
Hammers
HJ
,
Srinivas
S
, et al
Nivolumab versus everolimus in advanced renal-cell carcinoma
.
N Engl J Med
2015
;
373
:
1803
13
.
81.
Motzer
RJ
,
Tannir
NM
,
McDermott
DF
,
Arén Frontera
O
,
Melichar
B
,
Choueiri
TK
, et al
Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma
.
N Engl J Med
2018
;
378
:
1277
90
.
82.
Logothetis
CJ
,
Dexeus
FH
,
Finn
L
,
Sella
A
,
Amato
RJ
,
Ayala
AG
, et al
A prospective randomized trial comparing MVAC and CISCA chemotherapy for patients with metastatic urothelial tumors
.
J Clin Oncol
1990
;
8
:
1050
5
.
83.
Bellmunt
J
,
de Wit
R
,
Vaughn
DJ
,
Fradet
Y
,
Lee
J-L
,
Fong
L
, et al
Pembrolizumab as second-line therapy for advanced urothelial carcinoma
.
N Engl J Med
2017
;
376
:
1015
26
.
84.
Fradet
Y
,
Bellmunt
J
,
Vaughn
DJ
,
Lee
JL
,
Fong
L
,
Vogelzang
NJ
, et al
Randomized phase III KEYNOTE-045 trial of pembrolizumab versus paclitaxel, docetaxel, or vinflunine in recurrent advanced urothelial cancer: results of >2 years of follow-up
.
Ann Oncol Off J Eur Soc Med Oncol
2019
;
30
:
970
6
.
85.
Rosenberg
JE
,
Hoffman-Censits
J
,
Powles
T
,
van der Heijden
MS
,
Balar
AV
,
Necchi
A
, et al
Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial
.
Lancet
2016
;
387
:
1909
20
.
86.
Balar
AV
,
Galsky
MD
,
Rosenberg
JE
,
Powles
T
,
Petrylak
DP
,
Bellmunt
J
, et al
Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial
.
Lancet
2017
;
389
:
67
76
.
87.
Powles
T
,
Park
SH
,
Voog
E
,
Caserta
C
,
Valderrama
BP
,
Gurney
H
, et al
Maintenance avelumab + best supportive care (BSC) versus BSC alone after platinum-based first-line (1L) chemotherapy in advanced urothelial carcinoma (UC): JAVELIN Bladder 100 phase III interim analysis
.
J Clin Oncol
2020
;
38
:
LBA1
LBA1
.
88.
Powles
TB
,
Loriot
Y
,
Bellmunt
J
,
Sternberg
CN
,
Sridhar
S
,
Petrylak
DP
, et al
699O Avelumab first-line (1L) maintenance + best supportive care (BSC) vs BSC alone for advanced urothelial carcinoma (UC): association between clinical outcomes and exploratory biomarkers
.
Ann Oncol
2020
;
31
:
S552
3
.
89.
Alva
A
,
Csőszi
T
,
Ozguroglu
M
,
Matsubara
N
,
Geczi
L
,
Cheng
S.-S
, et al
LBA23 Pembrolizumab (P) combined with chemotherapy (C) vs C alone as first-line (1L) therapy for advanced urothelial carcinoma (UC): KEYNOTE-361
.
Ann Oncol
2020
;
31
:
S1155
.
90.
Ferris
RL
,
Blumenschein
G
,
Fayette
J
,
Guigay
J
,
Colevas
AD
,
Licitra
L
, et al
Nivolumab for recurrent squamous-cell carcinoma of the head and neck
.
N Engl J Med
2016
;
375
:
1856
67
.
91.
Cohen
EEW
,
Soulières
D
,
Le Tourneau
C
,
Dinis
J
,
Licitra
L
,
Ahn
M-J
, et al
Pembrolizumab versus methotrexate, docetaxel, or cetuximab for recurrent or metastatic head-and-neck squamous cell carcinoma (KEYNOTE-040): a randomised, open-label, phase 3 study
.
Lancet (London, England)
2019
;
393
:
156
67
.
92.
Burtness
B
,
Harrington
KJ
,
Greil
R
,
Soulières
D
,
Tahara
M
,
de Castro
G
, et al
Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): a randomised, open-label, phase 3 study
.
Lancet
2019
;
394
:
1915
28
.
93.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
94.
Shitara
K
,
Özgüroğlu
M
,
Bang
Y-J
,
Di Bartolomeo
M
,
Mandalà
M
,
Ryu
M-H
, et al
Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial
.
Lancet
2018
;
392
:
123
33
.
95.
El-Khoueiry
AB
,
Sangro
B
,
Yau
T
,
Crocenzi
TS
,
Kudo
M
,
Hsu
C
, et al
Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial
.
Lancet
2017
;
389
:
2492
502
.
96.
Brahmer
JR
,
Tykodi
SS
,
Chow
LQM
,
Hwu
W-J
,
Topalian
SL
,
Hwu
P
, et al
Safety and activity of anti-PD-L1 antibody in patients with advanced cancer
.
N Engl J Med
2012
;
366
:
2455
65
.
97.
Marabelle
A
,
Le
DT
,
Ascierto
PA
,
Di Giacomo
AM
,
De Jesus-Acosta
A
,
Delord
J-P
, 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
.
98.
Pagès
F
,
Mlecnik
B
,
Marliot
F
,
Bindea
G
,
Ou
F-S
,
Bifulco
C
, et al
International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study
.
Lancet
2018
;
391
:
2128
39
.
99.
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
.
100.
Andre
T
,
Shiu
K-K
,
Kim
TW
,
Jensen
BV
,
Jensen
LH
,
Punt
CJA
, et al
Pembrolizumab versus chemotherapy for microsatellite instability-high/mismatch repair deficient metastatic colorectal cancer: the phase 3 KEYNOTE-177 study
.
J Clin Oncol
2020
;
38
:
LBA4
LBA4
.
101.
Shitara
K
,
Van Cutsem
E
,
Bang
Y-J
,
Fuchs
C
,
Wyrwicz
L
,
Lee
K-W
, et al
Efficacy and safety of pembrolizumab or pembrolizumab plus chemotherapy vs chemotherapy alone for patients with first-line, advanced gastric cancer
.
JAMA Oncol
2020
;
6
:
1571
.
102.
Moehler
M
,
Shitara
K
,
Garrido
M
,
Salman
P
,
Shen
L
,
Wyrwicz
L
, et al
LBA6_PR Nivolumab (nivo) plus chemotherapy (chemo) versus chemo as first-line (1L) treatment for advanced gastric cancer/gastroesophageal junction cancer (GC/GEJC)/esophageal adenocarcinoma (EAC): first results of the CheckMate 649 study
.
Ann Oncol
2020
;
31
:
S1191
.
103.
Boku
N
,
Ryu
MH
,
Oh
D-Y
,
Oh
SC
,
Chung
HC
,
Lee
K-W
, et al
LBA7_PR Nivolumab plus chemotherapy versus chemotherapy alone in patients with previously untreated advanced or recurrent gastric/gastroesophageal junction (G/GEJ) cancer: ATTRACTION-4 (ONO-4538-37) study
.
Ann Oncol
2020
;
31
:
S1192
.
104.
Kelly
RJ
,
Ajani
JA
,
Kuzdzal
J
,
Zander
T
,
Van Cutsem
E
,
Piessen
G
, et al
LBA9_PR Adjuvant nivolumab in resected esophageal or gastroesophageal junction cancer (EC/GEJC) following neoadjuvant chemoradiation therapy (CRT): first results of the CheckMate 577 study
.
Ann Oncol
2020
;
31
:
S1193
4
.
105.
Kato
K
,
Sun
J-M
,
Shah
MA
,
Enzinger
PC
,
Adenis
A
,
Doi
T
, et al
LBA8_PR Pembrolizumab plus chemotherapy versus chemotherapy as first-line therapy in patients with advanced esophageal cancer: the phase 3 KEYNOTE-590 study
.
Ann Oncol
2020
;
31
:
S1192
3
.
106.
Singh
RR
,
O'Reilly
EM
. 
New treatment strategies for metastatic pancreatic ductal adenocarcinoma
.
Drugs
2020
;
80
:
647
69
.
107.
Renouf
DJ
,
Knox
JJ
,
Kavan
P
,
Jonker
D
,
Welch
S
,
Couture
F
, et al
LBA65 The Canadian Cancer Trials Group PA.7 trial: Results of a randomized phase II study of gemcitabine (GEM) and nab-paclitaxel (Nab-P) vs GEM, nab-P, durvalumab (D) and tremelimumab (T) as first line therapy in metastatic pancreatic ductal adenocarcinoma (mPDAC)
.
Ann Oncol
2020
;
31
:
S1195
.
108.
Bockorny
B
,
Semenisty
V
,
Macarulla
T
,
Borazanci
E
,
Wolpin
BM
,
Stemmer
SM
, et al
BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: the COMBAT trial
.
Nat Med
2020
;
26
:
878
85
.
109.
Voorwerk
L
,
Slagter
M
,
Horlings
HM
,
Sikorska
K
,
van de Vijver
KK
,
de Maaker
M
, et al
Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: the TONIC trial
.
Nat Med
2019
;
25
:
920
8
.
110.
Champiat
S
,
Dercle
L
,
Ammari
S
,
Massard
C
,
Hollebecque
A
,
Postel-Vinay
S
, et al
Hyperprogressive disease is a new pattern of progression in cancer patients treated by anti-PD-1/PD-L1
.
Clin Cancer Res
2017
;
23
:
1920
8
.
111.
Melero
I
,
Berraondo
P
,
Rodríguez-Ruiz
ME
,
Pérez-Gracia
JL
. 
Making the most of cancer surgery with neoadjuvant immunotherapy
.
Cancer Discov
2016
;
6
:
1312
4
.