Abstract
Clinicians caring for patients with sarcoma founded the field of cancer immunotherapy. Despite this, contemporary success with immunotherapy for sarcoma has been limited. Here, we review immunotherapy for sarcoma including Coley’s toxins, interleukin-2, adoptive cell transfer, and checkpoint blockade. We detail recent and ongoing efforts to combine checkpoint blockade with other immune modulators, surgery, or radiation. These results, along with ongoing investigations, have identified immunotherapeutic approaches as a promising avenue for progress in advanced sarcomas.
Keywords: bone sarcoma, soft tissue sarcoma, sarcomas, immunotherapy, checkpoint inhibition
INTRODUCTION
“The fact that in one of the reported cases when two attacks of erysipelas had accidentally occurred, the first diminishing the size of the tumor, and the second causing it to disappear, led me to try repeated inoculations.” – W. B. Coley, 18901
The first cancer immunotherapists were, famously, surgeons caring for patients with sarcoma. An important early advance was the discovery of the etiology of erysipelas by the 19th century German surgeon Friedrich Fehleisen, who showed it could be produced by inoculation with cultures of Streptococcus erysipelatis2. After reading reports of tumor regression in patients with sarcoma who developed erysipelas, William Coley then deliberately provoked erysipelas by inoculation1. In some cases, this resulted in dramatic tumor shrinkage.
The mechanism of tumor regression induced by erysipelas infection escaped Coley, but was later discovered by John J. Morton, a surgeon who would go on to serve in France under Harvey Cushing during the Great War. Morton showed that rejection of murine tumors was associated with infiltration of lymphocytes and concluded, “the lymphocyte is a necessary factor in cancer immunity.”3 Fifty years later, the British surgeon E. J. Delorme showed that adoptive transfer of lymphocytes in a mouse model could inhibit carcinogen-induced sarcoma growth4. Collectively, these findings helped give form to the notion that lymphocytes may be used therapeutically to treat patients with advanced sarcoma.
A breakthrough came in the 1970s with the discovery of interleukin-2 (IL-2), which enabled in vitro culture of T lymphocytes5,6. This paved the way for the modern era of immunotherapy, which began in 1988 when surgeon Steven A. Rosenberg and colleagues demonstrated adoptive cell transfer (ACT) of tumor infiltrating lymphocytes (TIL) could effectively treat advanced melanoma in studies conducted in the Surgery Branch of the National Cancer Institute (NCI)7. Subsequent trials showed high dose IL-2 in some patients induced durable regression of advanced renal cell carcinoma (RCC) and melanoma8,9. Rosenberg & colleagues made other key advances in the field of T cell therapy including the use of a preparative regimen of lymphodepleting chemotherapy, which enhances the engraftment and persistence of transferred T cells10. Surgery Branch investigators also showed that specific immune responses could be directed against a known cancer antigen by gene modification of autologous lymphocytes with a T cell receptor (TCR) of choice11.
A major distinction between immunotherapy and cytotoxic chemotherapy or targeted therapy for solid tumors in adults is that immune-mediated complete responses (CR) are usually durable for years or decades. For example, 27 (82%) of 33 patients with melanoma or renal cell carcinoma who had a CR after IL-2 were disease-free at a median of 86 months of follow-up12. Fourteen (93%) of 15 patients with melanoma who had a CR after CTLA-4 blockade (with or without IL-2) remained disease-free at a median follow-up of 83 months13. In two trials evaluating ACT of TIL for melanoma, CRs were observed in 22–24% of patients, and 90% of those responses were durable at long follow-up14,15. These results show the immune system is capable of sterilizing, or at least permanently suppressing, the entire burden of metastatic disease in some patients.
Sarcomas are a heterogeneous group of mesenchymal neoplasms of bone and soft tissue origin and include more than 70 distinct subtypes. Approximately 15,000 cases of soft tissue and bone sarcomas are diagnosed annually in the U.S. Despite primary combined modality therapy, 25–50% of patients develop recurrent and/or metastatic disease16,17. For patients with unresectable advanced disease, chemotherapy remains the standard of care. However, complete responses are rare and the median survival in the metastatic setting is 15–20 months18–21. Standard cytotoxic chemotherapy agents such as doxorubicin, ifosfamide, and dacarbazine result in objective responses in 10–25% of patients, but response rate varies significantly among histologic variants, and responses are typically not durable22. Therefore, novel and effective systemic therapies are desperately needed for patients with sarcomas.
INTERLEUKIN-2 FOR SARCOMA
There have been surprisingly few studies evaluating the use of IL-2 for patients with advanced sarcoma but results from one trial suggest it might be effective. Investigators enrolled ten pediatric patients with metastatic neuroblastoma, osteosarcoma, Ewing’s sarcoma or Wilms’ tumor23. Five (50%) of 10 patients had a CR to IL-2, and those responses were durable at a median follow-up of 28 months. The study investigators reported severe but reversible toxicities.
Despite proof of efficacy in melanoma and RCC, IL-2 is not widely used for those diseases. It is dosed every 8 hours in the inpatient setting, which requires a significant commitment on the part of providers. Furthermore, perceptions regarding IL-2 toxicity, and cytokine-induced capillary leak syndrome in particular, have dampened enthusiasm for the treatment24. It is worth mentioning that in experienced hands IL-2 toxicity can be manageable. One study reported 809 consecutively treated patients without a single treatment-related death25.
However, in order to mitigate challenges with dosing and toxicity associated with IL-2 administration, a PEGylated recombinant form of IL-2 (Bempegaldesleukin) has been developed that is dosed every 3 weeks and is well tolerated26. In one study, cytokine-related symptoms (flu-like symptoms, rash, pruritus, or hypotension) were observed primarily in cycles one and two and became significantly reduced with additional cycles. Also, there was no grade ≥3 hypotension following the implementation of hydration guidelines.
Early results from studies using Bempegaldesleukin for solid tumors are very promising. Data from the phase 1/2 PIVOT-02 study (NCT03635983) showed that Bempegaldesleukin plus nivolumab resulted in a 60% objective response rate for patients with melanoma, RCC and non-small cell lung cancer. For patients with advanced sarcoma, preliminary results of a study evaluating Bempegaldesleukin plus nivolumab were presented at ASCO in 2019, which we discuss in more detail below. More research is needed in order to determine whether non-specific immune stimulation using cytokines such as IL-2 have a role in helping to boost anti-tumor immune responses for sarcoma.
ADOPTIVE CELL TRANSFER FOR SARCOMA
Cell therapy using autologous TIL is effective in patients with melanoma, and has shown promise in other solid tumors including cervical cancer and gastrointestinal epithelial malignancies27–29. D’Angelo and colleagues showed that TIL are present in a substantial fraction of sarcomas30. While there are no reports to date of adoptive transfer of TIL for sarcoma, this approach is currently being evaluated in clinical trials at several institutions (NCT04052334, NCT03449108, NCT03935893). It is also being evaluated for commercial application by Iovance Biotherapeutics (Wardell et al. 2019 Society for ImmunoTherapy of Cancer Annual Meeting, National Harbor, MD USA).
It is possible that a relative paucity of immunogenic neoantigens will limit the efficacy of TIL for sarcoma31. Rather than relying on the endogenous anti-tumor specificity of TIL, an alternative cell therapy approach involves gene-modification of lymphocytes with a TCR directed against an antigen of choice in an HLA-matched patient. The potential for efficacy of such an approach has been demonstrated in synovial sarcoma. Synovial sarcoma is a translocation-associated sarcoma and has few known neoantigens; perhaps in consequence, it does not respond well to checkpoint inhibitors (discussed in more detail below). However, synovial cell sarcoma almost universally expresses the cancer-germline antigen NY-ESO-1, which is not expressed in healthy somatic tissues.
Surgery Branch, NCI investigators showed that adoptive transfer of NY-ESO-1 specific peripheral blood lymphocytes (in HLA-A02 patients) resulted in objective responses in 11 (61%) of 18 patients with synovial cell sarcomas32. Investigators at Memorial Sloan Kettering Cancer Center (MSKCC) also showed that adoptive transfer of NY-ESO-1 specific T cells resulted in objective responses in 6 (50%) of 12 patients with synovial cell sarcoma33. MSKCC investigators are currently evaluating this approach for patients with myxoid/round cell liposarcomas, which also express NY-ESO-134. There are dozens of other cancer germline antigens that could theoretically be targeted in this fashion.
A major challenge for TCR therapy is identifying suitable patients, who must express both the target antigen and be HLA-matched35. An alternative to TCR gene therapy, one that obviates the need for HLA matching, is chimeric antigen receptor (CAR) T cell therapy. CAR T cells have the advantage of not being restricted by HLA, but their disadvantage is that they cannot be used to target peptide-MHC complexes (and so cannot target intracellular peptides and most neoantigens). CAR T cells can however target many extracellular antigens, including glycoproteins36.
Of critical importance for both TCR and CAR T cell therapy is avoiding “off tumor, on target” toxicity. In hematological malignancies, CAR T cells can be used safely because the healthy B cells that express the target antigen (CD19 or CD20) are expendable. In contrast, many of the antigens expressed in solid tumors are also present in normal healthy tissues that are not expendable. In one dramatic example of this, adoptive transfer of lymphocytes targeting the cancer germline antigen MAGE-A3 resulted in fatality because the antigen was expressed at low levels in the brain37. In another case, adoptive transfer of anti-HER2 CAR T cells resulted in respiratory failure and death from cytokine storm triggered by low level recognition of ERBB2 on pulmonary epithelial cells38. In another study, anti-carcinoembryonic antigen CAR T cells mediated objective responses in patients with advanced colorectal cancer, but at the cost of severe dose-limiting colitis39.
Fortunately, there are tumor-exclusive cell surface antigens that can be targeted by CARs, and there are ways to engineer CAR T cells to enhance safety36,40. A recent study evaluated anti-HER2 CAR T cells for HER2+ sarcoma in a dose escalation and demonstrated safety, but there were no responses41. The subsequent addition of lymphodepletion improved T cell expansion and resulted in CRs in 2 (20%) of 10 patients treated42. The two CRs were in rhabdomyosarcoma and osteosarcoma, which are almost always refractory to checkpoint inhibitors; both responses are ongoing at 15 and 32 months.
Cell therapy has other important limitations. The preparative regimen of lymphodepleting chemotherapy can itself have side effects including neutropenia and result in prolonged hospitalization. Cell therapy requires significant expertise and costs associated with maintaining GMP (good manufacturing practice) facilities. Limitations specific to TIL include the need for surgical resection and variation in the quality of the T cell product43. Despite these limitations, cell therapy using TIL or gene-modified T cells is a promising avenue of investigation for patients with sarcoma.
CHECKPOINT BLOCKADE FOR SARCOMA
In general, sarcomas have a high rate of primary resistance to checkpoint blockade. However, in certain sarcoma subtypes there is a clear signal for efficacy with checkpoint inhibitors. In particular, alveolar soft part sarcomas (ASPS), vascular sarcomas (including angiosarcoma and epithelioid hemangioendothelioma) and undifferentiated pleomorphic sarcomas (UPS) appear to be sensitive to PD-1/PD-L1 blockade (Figure 1). Not surprisingly, the data also suggest combinatorial approaches to immune manipulation may increase response rates.
Figure 1. Objective response to PD-1/PD-L1 blockade in sarcoma.
All sarcoma patients were treated with PD-1 or PD-L1 blocking antibodies (nivolumab, pembrolizumab, durvalumab, atezolizumab) either as monotherapy or with another agent (ipilimumab, Bempegaldesleukin, tremelimumab, axitinib, metronomic cyclophosphamide, epacadostat, sunitinib, T-VEC). Response evaluations were by RECIST v1.1 (except Somaiah et al. 2020 ASCO, who reported responses by irRC). The dotted horizontal line indicates a 20% objective response rate. The eight histologies to the left had fewer than 20 patients total across all studies; the exact number of objective responses/number treated is shown above the bar. The ten middle histologies (from Ewing sarcoma to ASPS) had more than 20 treated patients reported in the literature. The bar graph shows mean with standard deviation. Each triangle represents 6–22 patients from a single study or aggregated from multiple studies with small numbers. The 39 liposarcoma and 40 UPS patients from the SARC028 expansion cohort were split into two groups in order to reflect the weight of those large samples. To enable comparison with non-sarcoma histologies, we included data from trials evaluating anti-PD-1 or anti-PD-L1 for patients with sun-shielded melanomas (acral, mucosal or uveal), cutaneous melanoma, and Merkel cell carcinoma. Vascular sarcoma includes angiosarcoma and epithelioid hemangioendothelioma. Abbreviations: GIST (gastrointestinal stromal tumor), NOS (not otherwise specified), UPS/MFH (undifferentiated pleomorphic sarcoma / malignant fibrous histiocytoma), ASPS (alveolar soft part sarcoma).
Single-agent checkpoint inhibition
Several studies have evaluated the use of antibodies against CTLA-4, PD-1 or PD-L1 as monotherapy for sarcomas, and in general the objective response rate is very low. A pilot study of ipilimumab (anti-CTLA-4) for synovial cell sarcoma resulted in no objective responses44. In 22 patients with sarcoma who were treated with ipilimumab in a phase I clinic, there were no objective responses45.
SARC028 investigators were the first to evaluate PD-1/PD-L1 axis blockade for patients with sarcoma. Pembrolizumab (anti-PD-1) was used in a two-cohort, single-arm open-label phase II trial46. Data from the SARC028 expansion cohort were recently presented at ASCO47. Responses in UPS were observed in 9 (23%) of 40 patients, two of which were CRs (by RECIST v1.1). Responses in liposarcoma (LPS) were less frequent, occurring in 4 (10%) of 39 patients.
The Alliance A091401 study was an open-label non-comparative randomized phase II study evaluating nivolumab (anti-PD-1) with or without ipilimumab for patients with metastatic sarcoma48. In the nivolumab monotherapy arm, 2 (5%) of 38 patients had objective responses. The responders had ASPS and leiomyosarcoma (LMS); a 3rd patient with sarcoma not otherwise specified (NOS) also had an unconfirmed partial response. In the Alliance A091401 expansion cohort, nivolumab mediated responses in 0 (0%) of 9 patients with gastrointestinal stromal tumor (GIST), 1 (8%) of 12 patients with de-differentiated LPS and 1 (8%) of 12 with UPS49.
A phase II trial evaluating nivolumab in 12 patients with uterine LMS reported no objective responses50. Results of a randomized trial for patients with GIST were recently reported, but no objective responses were seen in the nivolumab arm51. However, a phase II study of atezolizumab (anti-PD-L1) for ASPS reported 8 (42%) of 19 patients had a partial response (O’Sullivan Coyne et al. 2018 CTOS, Rome, Italy). These data show that, with the exception of UPS and ASPS, the clinical activity of single-agent checkpoint blockade in sarcoma is very low.
Dual checkpoint inhibition
In the nivolumab plus ipilimumab arm of the Alliance A091401 study, 6 (16%) of 38 evaluable patients had an objective response48. A seventh patient had an unconfirmed response. In the nivolumab plus ipilimumab arm of the Alliance trial expansion cohort, responses occurred in 0 (0%) of 9 GIST, 2 (14%) of 12 de-differentiated LPS and 2 (14%) of 12 UPS49. In a randomized trial evaluating immunotherapy for GIST, nivolumab plus ipilimumab mediated an objective response in 1 (8%) of 12 patients51. A phase II study evaluating durvalumab (anti-PD-L1) and tremelimumab (anti-CTLA-4) resulted in objective responses in 5 (50%) of 10 ASPS52. While limited by small numbers, these results highlight the potential importance of combinatorial immunotherapy strategies and sarcoma subtype selection for the future design of immunotherapy trials for sarcoma.
Checkpoint inhibition plus novel immunomodulatory approaches
Results of a phase II study evaluating pembrolizumab with the IDO1 inhibitor epacadostat were recently reported at ASCO53. Among 29 evaluable patients, 1 PR (3%) was observed in a patient with LMS. Given the negative results of the recently published Keynote-252 study, which evaluated epacadostat for patients with melanoma, it is unclear whether IDO1 inhibition enhances anti-PD-1 therapy for solid tumors54.
Talimogene laherparepvec (T-VEC) is an oncolytic immunotherapy derived from a modified human herpes virus designed to self-replicate within and lyse tumor cells, thereby releasing tumor antigens and promoting regional and systemic antitumor immunity. In a phase II study of pembrolizumab plus T-VEC for 20 patients with locally advanced or metastatic sarcoma, the response rate was 35%, with a median duration of response of 56 weeks55. Of note, 2 of 3 patients with angiosarcoma and 2 of 2 with UPS had objective responses. Other responses were seen in myxofibrosarcoma (MFS), sarcoma NOS and epithelioid sarcoma. Expansion cohorts in these subtypes (except sarcoma NOS) are now open (NCT03069378), and there is also a neoadjuvant cohort for high-risk UPS/MFS.
Bempegaldesleukin, as mentioned previously, is an experimental PEGylated form of IL-2 that activates and expands natural killer and CD8+ T cells via CD122 agonist activity. The combination of Bempegaldesleukin and nivolumab in melanoma showed impressive efficacy with a favorable toxicity profile. Early results from an ongoing study evaluating Bempegaldesleukin plus nivolumab for patients with sarcoma was recently presented at ASCO56. Objective responses were seen in 5 (9%) of 57 patients, including vascular sarcoma, LMS, UPS, and chondrosarcoma. Final results of the trial are still pending.
Checkpoint inhibition plus chemotherapy
A phase II study evaluated pembrolizumab with metronomic dosing of the lymphotoxic agent cyclophosphamide for patients with LMS, UPS, GIST and a few other sarcomas. An objective response occurred in 1 (2%) of 50 patients treated, in a patient with a solitary fibrous tumor57. It is unclear whether the combination of systemic chemotherapy with checkpoint inhibitors will be effective for patients with sarcoma.
Checkpoint inhibitors combined with TKI
The first study evaluating a combination of checkpoint inhibitors with tyrosine kinase inhibitors (TKI) in patients with sarcoma used ipilimumab with dasatinib for twenty patients with advanced GIST along with eight patients with other sarcomas58. There were no objective responses in the study. This result may reflect the choice of TKI in a heavily pre-treated GIST population. Whether this strategy can be effective for TKI-sensitive patients remains to be seen.
The ASPL-TFE3 fusion is universally present in ASPS and leads to HIF-1α accumulation and upregulation of proangiogenic factors59. This was the rationale behind a phase II study of pembrolizumab with the VEGF receptor TKI axitinib for patients with advanced sarcomas including ASPS. The combination of pembrolizumab and axitinib mediated objective responses in 6 (55%) of 11 ASPS, 1 of 6 LMS and 1 of 1 epithelioid sarcomas60. Early results from the phase I/II Immunosarc study, evaluating nivolumab plus the multi-TKI inhibitor sunitinib, showed an objective response rate of 11% (Martin Broto et al. 2020 ESMO Sarcoma & GIST Symposium, Milan Italy).
Biomarkers and Response to Immunotherapy
A major limitation of the current literature is that individual subtypes of sarcomas are quite rare, so there are few data with which to estimate the comparative effectiveness of checkpoint inhibitor treatment. Furthermore, PFS in patients with sarcomas treated with checkpoint inhibitors is brief, typically lasting 2–4 months, which reflects the fact that in most patients, the natural history of the disease is not detectably altered.
It should be noted that many of the results represented in Figure 1 were from studies that combined PD-1/PD-L1 blockade with other agents including ipilimumab, T-VEC, Bempegaldesleukin, et cetera. Nonetheless, responses are being consistently observed for certain histological subtypes of sarcoma. ASPS, vascular sarcomas and UPS/MFS have response rates that are near or above 20%. Evidence of activity also exists in de-differentiated LPS, although response rates for now are below 10%. There are also promising data from subtypes such as dedifferentiated chondrosarcoma and epithelioid sarcoma.
There is a strong correlation between tumor mutational burden (TMB) and histology-specific response (among all solid tumors) to PD-1 blocking antibodies61. This may be explained by the ability of endogenous T cells to recognize the gene products of cancer-specific mutations (neoantigens)29,62. A high TMB thus leads to an abundance of neoantigens which can be targeted by the immune system31. This hypothesis also explains divergent response patterns within some types of solid tumors. For example, cutaneous melanoma, which has a large burden of neoantigens due to UV mutagenesis, is highly sensitive to checkpoint blockade. In contrast, sun-shielded melanomas including acral lentiginous, mucosal or uveal melanomas have a paucity of neoantigens and are less responsive63. Mismatch repair-proficient colorectal cancer has a low TMB and is unresponsive to anti-PD-1, but mismatch repair-deficient colorectal cancer has a high TMB and is highly sensitive60.
Sarcomas in general are associated with a low TMB. One study used 100,000 human cancer genomes and ranked 167 human tumors by TMB, showing the most highly mutated sarcoma (angiosarcoma) was only ranked 67th overall64. Interestingly, the three sarcoma subtypes with the highest percentage of cases with more than 20 mutations per megabase were angiosarcoma (13.4% of cases), MPNST (8.2%) and UPS (8.1%). These subtypes are typically characterized by complex genotypes and often present in older patients. For comparison, only 0.5% of Ewing sarcomas and 0.2% of liposarcoma had more than 20 mutations per megabase64. Synovial sarcoma, which ranked 143rd among the tumors analyzed, is a translocation-associated sarcoma that usually results from the formation of SS18-SSX fusion oncogenes65. Consistent with its low TMB, only two responses to checkpoint blockade in patients with synovial sarcoma have been reported in the literature (Figure 1). ASPS was not included in the study, but also has a very low TMB as it is translocation-associated.
It is unclear whether PD-L1 expression is a useful biomarker in sarcoma. D’Angelo et al. showed that lymphocyte and macrophage infiltration is common in sarcoma, but PD-L1 expression is uncommon in most sarcomas30. Furthermore, while PD-L1 expression is high in GIST (present in 29% of cases), GIST has a very low response rate to immunotherapy thus far (Figure 1). There has been no observed association between PD-L1 expression and overall survival. It is worth noting that in the SARC028 study, 3 (4%) of 70 evaluated tumor samples had PD-L1 positivity at the 1% threshold, and 2 of those 3 had objective responses to treatment46. Also, Wilky et al. showed 11 of 11 ASPS tumor samples were PD-L1 positive60.
The metabolism connection
There are several curious similarities between clear cell RCC and ASPS. Clear cell RCC is as sensitive to IL-2 and PD-1 blockade as melanoma but has a low TMB66. RCC is characterized by mutations in VHL or other enzymes of metabolism that result in HIF accumulation66. ASPS is a rare sarcoma of uncertain mesenchymal origin that was first described at MSKCC in 195267. Of all sarcoma histological subtypes, ASPS appears to be the most responsive to immunotherapy (Figure 1). ASPS is a translocation-associated sarcoma and, like RCC, has a low TMB68.
Furthermore, it has been shown that the unbalanced ASPL-TFE3 fusion associated with ASPS leads to accumulation of HIF via dysregulated lactate metabolism59. Intriguingly, the ASPL-TFE3 fusion found in ASPS, which arises from an unbalanced translocation, can lead to primary renal neoplasms that resemble RCC if instead the fusion results from a balanced translocation69. Thus, clear cell RCC and ASPS share a low TMB, high response rates to immunotherapy, and are characterized by metabolic dysregulation.
It is possible that the metabolic phenotype of RCC and ASPS leads to enhanced immune infiltration of tumors. A recent study evaluated gene expression from 608 tumors across a variety of subtypes of soft tissue sarcoma (but did not include ASPS)70. Those data were used to identify distinct sarcoma immune classes (SIC): immune-low, immune high, and vascularized. The immune low SIC had low densities of CD3+, CD8+ or CD20+ cells. In contrast, the vascularized SIC showed a moderate infiltration of immune cells and a high density of CD34+ endothelial cells. RCC and ASPS upregulate angiogenesis signals via HIF, which may promote immune cell infiltration into the tumors, where they are poised to mediate regression.
Immunotherapy strategies in the surgical patient
The curative potential of immunotherapy provides clear rationale for a complementary role of local therapy in selected cases. After all, if an immune-mediated CR is usually durable, then a near-CR followed by local therapy to extirpate any remaining disease might also achieve durable disease control. Retrospective data support this conclusion, including two studies where patients with metastatic melanoma were treated with immunotherapy and then metastasectomy for oligoprogressive immunorefractory disease71,72. In some cases, patients were disease-free for years after surgery.
The rationale for a complementary relationship between immunotherapy and local therapy that has emerged in melanoma is largely predicated on a high frequency of robust responses. In a patient with unresectable metastatic melanoma, immunotherapy can alter the natural history of the disease and achieve systemic control, while isolated local failures are controlled with surgery or local therapy. As the efficacy of immunotherapy improves, this strategy might become more widely applicable, as patients with unresectable disease undergo “oligometastatic conversion” by immunotherapy. The potential for conversion to resectability depends on strong anti-tumor immune responses, which are frequently seen in melanoma. In sarcoma, this approach may not be useful until response rates to immunotherapy can be improved.
More exciting than the prospect of a complementary relationship between immunotherapy and locoregional therapy is the possibility of a synergistic relationship between the two. In one study, 17 matched UPS samples taken from before and after radiotherapy showed a trend towards increased density of TIL73. These and other data provide rationale for combination neoadjuvant immune checkpoint inhibition and radiotherapy. This approach is currently being evaluated in the SARC032 study, a phase II trial comparing neoadjuvant Pembrolizumab plus radiotherapy versus radiotherapy alone for patients with sarcoma (NCT03092323).
Investigators at MD Anderson are evaluating neoadjuvant checkpoint blockade with nivolumab or nivolumab plus ipilimumab in patients with surgically resectable retroperitoneal de-differentiated LPS or with extremity/truncal UPS, treated with concurrent neoadjuvant radiation therapy74. In a recent presentation, the median pathologic response in the UPS cohort was reported to be 95% and was similar between the two arms. Median pathologic response in the de-differentiated LPS cohort was more modest, at 22.5%. There were 8 patients with a pathological response ≥ 85%; 1 had a partial response, 5 had stable diseases and 2 had progressive disease.
At MSKCC, encouraging results from the phase II trial of pembrolizumab and T-VEC55 have led to an expansion cohort that includes neoadjuvant immunotherapy for patients with UPS/MFS (NCT03069378). Additionally, in the setting of locally advanced disease, we are employing a strategy of local chemotherapy (with melphalan and dactinomycin) administered by isolated limb infusion to increase the response rate of pembrolizumab (NCT04332874). MSKCC investigators previously demonstrated in patients with melanoma that a combination of high-dose chemotherapy with checkpoint inhibition promoted an inflammatory response and sensitized the cancer both locally and systemically75. In forthcoming trials in sarcoma, correlative studies will be aimed at characterizing the pretreatment sarcoma tumor microenvironment and changes to the immune infiltrate induced by the combinatorial approaches.
Current Clinical Context
Immunotherapy in the form of checkpoint inhibition should be a consideration in patients with inoperable locally advanced/metastatic sarcoma with specific subtypes including UPS, angiosarcoma, and ASPS. The optimal time to introduce immunotherapy in the systemic therapy treatment paradigm for these sarcoma subtypes remains unknown. In other disease settings, inferior response rates to immunotherapy have been observed when administered after chemotherapy compared to when introduced in the first line setting76,77. Hence, consideration of immunotherapy earlier in patients with advanced UPS, angiosarcoma, or ASPS is reasonable.
Combinatorial therapies may ultimately prove more efficacious, but many trials testing these strategies exclude patients with prior checkpoint inhibition. For this reason, a review of available clinical trials should be performed before starting immunotherapy for metastatic sarcoma. Trials evaluating immunotherapy for earlier stage disease hold the promise of reduced toxicity and/or potentially improved efficacy in the (neo)adjuvant setting but are currently limited to patients at highest risk of recurrence with histologic subtypes thought to be most likely to benefit. The field of immunotherapy is rapidly evolving, and individual sarcoma histologic types are rare. In this setting, smaller, hypothesis-driven trials in homogeneous patient cohorts are likely to provide the most efficient way to identify particularly promising new approaches for patients with sarcoma.
Conclusions
Clinicians treating patients with sarcomas pioneered the field of cancer immunotherapy. Recent advances have most benefited patients whose solid tumors have a high tumor mutational burden, but there is abundant evidence showing immunotherapy can successfully treat certain neoantigen-sparse malignancies. Immunotherapy is most promising in ASPS, vascular sarcoma and UPS. Dual checkpoint inhibitor or combinatorial immune modulation may result in higher response rates. Multidisciplinary management with locoregional approaches and perhaps cancer-specific targeting are likely to be indispensable for clinical success. Biomarkers to predict response and improve patient selection will further refine clinical algorithms, but at present these are largely absent. It has been over 100 years since Coley first unleashed the immune system against sarcomas, but it does appear that immunotherapy for sarcoma may finally be coming of age.
Methods for Figure 1
We included data from published trials evaluating PD-1 or PD-L1 antibodies either as monotherapy or in combination with another agent (cyclophosphamide, axitinib, ipilimumab, epacadostat or T-VEC)46,48,57 50,53,60. We also included data from unpublished trials evaluating PD-1 or PD-L1 antibodies with or without other agents that were presented at national meetings, including:
A phase II study of atezolizumab in patients with alveolar soft part sarcoma (O’Sullivan Coyne et al. CTOS 2018 Annual Meeting, Rome, Italy)
A randomized phase II study of nivolumab monotherapy versus nivolumab combined with ipilimumab in advanced gastrointestinal stromal tumor (Singh et al. 2019 American Society of Clinical Oncology, Chicago, USA)
SARC028 expansion cohort (Burgess et al. 2019 American Society of Clinical Oncology, Chicago, USA)
A phase II study evaluating Durvalumab plus Tremelimumab (Somaiah et al. 2020 American Society of Clinical Oncology, Chicago, USA)
A multicenter phase II study of nivolumab +/− ipilimumab for patients with metastatic sarcoma (Alliance A091401): Results of expansion cohorts (Chen et al. 2020 American Society of Clinical Oncology, Chicago, USA)
A phase II study evaluating Nivolumab plus Bempegaldesleukin (D’Angelo et al. 2019 American Society of Clinical Oncology, Chicago, USA).
A phase I/II Immunosarc study evaluating Nivolumab plus sunitinib (Martin Broto et al. 2020 European Society for Medical Oncology Sarcoma & GIST Symposium, Milan, Italy)
For those histologies for which fewer than 20 treated patients were reported in the literature (spindle cell sarcoma to myxofibrosarcoma), all patients were aggregated into a single group and presented as a bar graph. No standard deviation is shown. Above the bar shows the exact number of objective responses (by RECIST v1.1, except the study by Somaiah et al. which used irRC) and the number of patients treated.
For those histologies with more than 20 patients treated in the literature (Ewing sarcoma to ASPS), the bar graph shows mean with standard deviation. Each triangle represents 6–22 patients from a single study or aggregated from multiple studies with small numbers (1–5 patients each). The 39 liposarcoma and 40 UPS patients from the SARC028 expansion cohort are represented by two triangles in order to reflect the weight of those samples.
For purposes of comparison, data from trials evaluating anti-PD-1 or anti-PD-L1 monotherapy for patients with sun-shielded melanoma (1 acral, 2 mucosal, 2 uveal), cutaneous melanoma and Merkel cell carcinoma are shown on the far right of Figure 1. Response rates for sun-shielded melanomas include: 23.3% for mucosal melanoma and 40.9% for cutaneous melanoma78, 32% for acral melanoma and 23% for mucosal79, and 3.6% and 4.7% for uveal melanoma80,81.
Trials evaluating PD-1/PD-L1 monotherapy for cutaneous melanoma reported an ORR of 28% (NCT00730639)82, an ORR of 44.3% (NCT01844505, CheckMate067)83, and ORR of 33.7% and 32.9% (NCT01866319, KEYNOTE-066)84. Trials evaluating PD-1 / PD-L1 for Merkel cell carcinoma included an ORR of 56% for Pembrolizumab (NCT02267603)85, ORR of 31.8% for Avelumab (NCT02155647)77, and ORR of 68% for Nivolumab (NCT02488759; CheckMate 358) (Topalian et al. 2017 American Association for Cancer Research Annual Meeting, Washington DC).
Data Availability
Data generated for this manuscript are available from the corresponding author upon reasonable request.
SYNOPSIS.
Sarcomas as a class have been largely refractory to immunotherapy. However, emerging data show promising evidence of efficacy of immune-based approaches, especially for certain subtypes of sarcoma.
Sources of Support:
This research is funded in part by a grant from the National Cancer Institute / National Institutes of Health (P30-CA008748).
Commercial Interests:
Dr. Kelly reports receiving research funding from Amgen, Merck, Kartos, Exicure and Agios and serves as a consultant for Exicure. Dr. Klemen and Bartlett have no relevant conflicts of interest to report
REFERENCES
- 1.Coley WB. II. Contribution to the Knowledge of Sarcoma. Annals of Surgery 1891;14(3):199–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Fehleisen F Die Aetiologie des Erysipels. 1883. [Google Scholar]
- 3.Murphy JB, Morton JJ. The Lymphocyte as a Factor in Natural and Induced Resistance to Transplanted Cancer. Proc Natl Acad Sci USA 1915;1(7):435–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.DELORME EJ, ALEXANDER P. TREATMENT OF PRIMARY FIBROSARCOMA IN THE RAT WITH IMMUNE LYMPHOCYTES. The Lancet 1964;2(7351):117–20. [DOI] [PubMed] [Google Scholar]
- 5.Morgan DA, Ruscetti FW, Gallo R. Selective in vitro growth of T lymphocytes from normal human bone marrows. Science 1976;193(4257):1007–8. [DOI] [PubMed] [Google Scholar]
- 6.Ruscetti FW, Morgan DA, Gallo RC. Functional and morphologic characterization of human T cells continuously grown in vitro. The Journal of Immunology 1977;119(1):131–8. [PubMed] [Google Scholar]
- 7.Rosenberg SA, Packard BS, Aebersold PM. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. The New England … 1988;319(25):1676–80. [DOI] [PubMed] [Google Scholar]
- 8.Atkins MB, Lotze MT, Dutcher JP, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. JCO 1999;17(7):2105–16. [DOI] [PubMed] [Google Scholar]
- 9.Rosenberg SA. Raising the bar: the curative potential of human cancer immunotherapy. Sci Transl Med 2012;4(127):127ps8–127ps8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Dudley ME, Wunderlich JR, Yang JC, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. JCO 2005;23(10):2346–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Morgan RA, Dudley ME, Wunderlich JR, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 2006;314(5796):126–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Rosenberg SA, Yang JC, White DE, Steinberg SM. Durability of complete responses in patients with metastatic cancer treated with high-dose interleukin-2: identification of the antigens mediating response. Annals of Surgery 1998;228(3):307–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Prieto PA, Yang JC, Sherry RM, et al. CTLA-4 blockade with ipilimumab: long-term follow-up of 177 patients with metastatic melanoma. Clinical Cancer Research 2012;18(7):2039–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Rosenberg SA, Yang JC, Sherry RM, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clinical Cancer Research 2011;17(13):4550–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Goff SL, Dudley ME, Citrin DE, 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;:JCO667220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lindberg RD, Martin RG, Romsdahl MM, Barkley HT. Conservative surgery and postoperative radiotherapy in 300 adults with soft-tissue sarcomas. Cancer 1981;47(10):2391–7. [DOI] [PubMed] [Google Scholar]
- 17.Weitz J, Antonescu CR, Brennan MF. Localized extremity soft tissue sarcoma: improved knowledge with unchanged survival over time. JCO 2003;21(14):2719–25. [DOI] [PubMed] [Google Scholar]
- 18.Billingsley KG, Burt ME, Jara E, et al. Pulmonary metastases from soft tissue sarcoma: analysis of patterns of diseases and postmetastasis survival. Annals of Surgery 1999;229(5):602–10–discussion610–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Tap WD, Wagner AJ, Schöffski P, et al. Effect of Doxorubicin Plus Olaratumab vs Doxorubicin Plus Placebo on Survival in Patients With Advanced Soft Tissue Sarcomas: The ANNOUNCE Randomized Clinical Trial. JAMA 2020;323(13):1266–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.PhD DBS, FRCP SJS, MD PJW, et al. Articles Gemcitabine and docetaxel versus doxorubicin as rst-line treatment in previously untreated advanced unresectable or metastatic soft-tissue sarcomas (GeDDiS): a randomised controlled phase 3 trial. Lancet Oncology 2017;18(10):1397–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Van Glabbeke M, van Oosterom AT, Oosterhuis JW, et al. Prognostic factors for the outcome of chemotherapy in advanced soft tissue sarcoma: an analysis of 2,185 patients treated with anthracycline-containing first-line regimens--a European Organization for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group Study. JCO 1999;17(1):150–7. [DOI] [PubMed] [Google Scholar]
- 22.Brennan MF, Antonescu CR, Maki RG. Management of soft tissue sarcoma. 2013. [Google Scholar]
- 23.Schwinger W, Klass V, Benesch M, et al. Feasibility of high-dose interleukin-2 in heavily pretreated pediatric cancer patients. Annals of Oncology 2005;16(7):1199–206. [DOI] [PubMed] [Google Scholar]
- 24.Rosenberg SA. IL-2: the first effective immunotherapy for human cancer. J Immunol 2014;192(12):5451–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kammula US, White DE, Rosenberg SA. Trends in the safety of high dose bolus interleukin-2 administration in patients with metastatic cancer. Cancer 1998;83(4):797–805. [PubMed] [Google Scholar]
- 26.Diab A, Tannir NM, Bentebibel S-E, 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(8):1158–73. [DOI] [PubMed] [Google Scholar]
- 27.Stevanović S, Draper LM, Langhan MM, et al. Complete regression of metastatic cervical cancer after treatment with human papillomavirus-targeted tumor-infiltrating T cells. J Clin Oncol 2015;33(14):1543–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Tran E, Turcotte S, Gros A, et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 2014;344(6184):641–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Tran E, Ahmadzadeh M, Lu Y-C, et al. Immunogenicity of somatic mutations in human gastrointestinal cancers. Science 2015;350(6266):1387–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.D’Angelo SP, Shoushtari AN, Agaram NP, et al. Prevalence of tumor-infiltrating lymphocytes and PD-L1 expression in the soft tissue sarcoma microenvironment. Human Pathology 2015;46(3):357–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lawrence MS, Stojanov P, Polak P, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 2013;499(7457):214–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Robbins PF, Kassim SH, Tran TLN, et al. A Pilot Trial Using Lymphocytes Genetically Engineered with an NY-ESO-1-Reactive T-cell Receptor: Long-term Follow-up and Correlates with Response. Clinical Cancer Research 2015;21(5):1019–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.D’Angelo SP, Melchiori L, Merchant MS, et al. Antitumor Activity Associated with Prolonged Persistence of Adoptively Transferred NY-ESO-1 c259T Cells in Synovial Sarcoma. Cancer Discov 2018;8(8):944–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.D’Angelo SP, Druta M, Liebner DA, et al. Pilot study of NY-ESO-1c259 T cells in advanced myxoid/round cell liposarcoma. JCO 2018;36(15_suppl):3005–5. [Google Scholar]
- 35.Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015;348(6230):62–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Louis CU, Savoldo B, Dotti G, et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 2011;118(23):6050–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Morgan RA, Chinnasamy N, Abate-Daga D, et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother 2013;36(2):133–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case Report of a Serious Adverse Event Following the Administration of T Cells Transduced With a Chimeric Antigen Receptor Recognizing ERBB2. Mol Ther 2010;18(4):843–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Parkhurst MR, Yang JC, Langan RC, et al. T Cells Targeting Carcinoembryonic Antigen Can Mediate Regression of Metastatic Colorectal Cancer but Induce Severe Transient Colitis. Molecular Therapy 2009;19(3):620–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Brown CE, Alizadeh D, Starr R, et al. Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. N Engl J Med 2016;375(26):2561–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Ahmed N, Brawley VS, Hegde M, et al. Human Epidermal Growth Factor Receptor 2 (HER2) -Specific Chimeric Antigen Receptor-Modified T Cells for the Immunotherapy of HER2-Positive Sarcoma. J Clin Oncol 2015;33(15):1688–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Navai SA, Derenzo C, Joseph S, et al. Abstract LB-147: Administration of HER2-CAR T cells after lymphodepletion safely improves T cell expansion and induces clinical responses in patients with advanced sarcomas. Cancer Res 2019;79(13 Supplement):LB–147–LB–147. [Google Scholar]
- 43.Crompton JG, Klemen N, Kammula US. Metastasectomy for Tumor-Infiltrating Lymphocytes: An Emerging Operative Indication in Surgical Oncology. Ann Surg Oncol 2017;12(20):269–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Maki RG, Jungbluth AA, Gnjatic S, et al. A Pilot Study of Anti-CTLA4 Antibody Ipilimumab in Patients with Synovial Sarcoma. Sarcoma 2013;2013(22):1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Groisberg R, Hong DS, Behrang A, et al. Characteristics and outcomes of patients with advanced sarcoma enrolled in early phase immunotherapy trials. 2017;:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Tawbi HA, Burgess M, Bolejack V, et al. Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial. The Lancet Oncology 2017;18(11):1493–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Burgess MA, Bolejack V, Schuetze S, et al. Clinical activity of pembrolizumab (P) in undifferentiated pleomorphic sarcoma (UPS) and dedifferentiated/pleomorphic liposarcoma (LPS): Final results of SARC028 expansion cohorts. JCO 2019;37(15_suppl):11015–5. [Google Scholar]
- 48.D’Angelo SP, Mahoney MR, Van Tine BA, et al. Nivolumab with or without ipilimumab treatment for metastatic sarcoma (Alliance A091401): two open-label, non-comparative, randomised, phase 2 trials. The Lancet Oncology 2018;19(3):416–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chen JL, Mahoney MR, George S, et al. A multicenter phase II study of nivolumab +/− ipilimumab for patients with metastatic sarcoma (Alliance A091401): Results of expansion cohorts. JCO 2020;38(15_suppl):11511–1. [Google Scholar]
- 50.Ben-Ami E, Barysauskas CM, Solomon S, et al. Immunotherapy with single agent nivolumab for advanced leiomyosarcoma of the uterus: Results of a phase 2 study. Cancer 2017;123(17):3285–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Singh AS, Chmielowski B, Hecht JR, et al. A randomized phase II study of nivolumab monotherapy versus nivolumab combined with ipilimumab in advanced gastrointestinal stromal tumor (GIST). JCO 2019;37(15_suppl):11017–7. [Google Scholar]
- 52.Somaiah N, Conley AP, Lin HY, et al. A phase II multi-arm study of durvalumab and tremelimumab for advanced or metastatic sarcomas. JCO 2020;38(15_suppl):11509–9. [Google Scholar]
- 53.Kelly CM, Chi P, Dickson MA, Gounder MM. A phase II study of epacadostat and pembrolizumab in patients with advanced sarcoma. JCO 2019;37(15_suppl)11049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Long GV, Dummer R, Hamid O, 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. The Lancet Oncology 2019;20(8):1083–97. [DOI] [PubMed] [Google Scholar]
- 55.Kelly CM, Antonescu CR, Bowler T, et al. Objective Response Rate Among Patients With Locally Advanced or Metastatic Sarcoma Treated With Talimogene Laherparepvec in Combination With Pembrolizumab. JAMA Oncol 2020;6(3):402–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.D’Angelo SP, Conley AP, Kelly CM, et al. Pilot study of NKTR214 and nivolumab in patients with sarcomas. JCO 2019;37(15_suppl):11010–0. [Google Scholar]
- 57.Toulmonde M, Penel N, Adam J, et al. Use of PD-1 Targeting, Macrophage Infiltration, and IDO Pathway Activation in Sarcomas. JAMA Oncol 2018;4(1):93–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.D’Angelo SP, Shoushtari AN, Keohan ML, et al. Combined KIT and CTLA-4 Blockade in Patients with Refractory GIST and Other Advanced Sarcomas: A Phase Ib Study of Dasatinib plus Ipilimumab. Clinical Cancer Research 2017;23(12):2972–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Goodwin ML, Jin H, Straessler K, et al. Modeling alveolar soft part sarcomagenesis in the mouse: a role for lactate in the tumor microenvironment. Cancer Cell 2014;26(6):851–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Wilky BA, Trucco MM, Subhawong TK, et al. Axitinib plus pembrolizumab in patients with advanced sarcomas including alveolar soft-part sarcoma: a single-centre, single-arm, phase 2 trial. The Lancet Oncology 2019;20(6):837–48. [DOI] [PubMed] [Google Scholar]
- 61.Yarchoan M, Hopkins A, Jaffee EM. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N Engl J Med 2017;377(25):2500–1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Robbins PF, Lu Y-C, El-Gamil M, et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med 2013;19(6):747–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Klemen ND, Wang M, Rubinstein JC, et al. Survival after checkpoint inhibitors for metastatic acral, mucosal and uveal melanoma. Journal for ImmunoTherapy of Cancer 2013 1:1 2020;8(1):e000341–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med 2017;9(1):34–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Thway K, Fisher C. Synovial sarcoma: defining features and diagnostic evolution. Annals of Diagnostic Pathology 2014;18(6):369–80. [DOI] [PubMed] [Google Scholar]
- 66.The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 2013;499(7456):43–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.CHRISTOPHERSON WM, FOOTE FW, STEWART FW. Alveolar soft-part sarcomas; structurally characteristic tumors of uncertain histogenesis. Cancer 1952;5(1):100–11. [DOI] [PubMed] [Google Scholar]
- 68.Groisberg R, Roszik J, Conley AP, et al. Genomics, Morphoproteomics, and Treatment Patterns of Patients with Alveolar Soft Part Sarcoma and Response to Multiple Experimental Therapies. Mol Cancer Ther 2020;19(5):1165–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Argani P, Antonescu CR, Illei PB, et al. Primary renal neoplasms with the ASPL-TFE3 gene fusion of alveolar soft part sarcoma: a distinctive tumor entity previously included among renal cell carcinomas of children and adolescents. Am J Pathol 2001;159(1):179–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Petitprez F, AXLRX s, Keung EZ, et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 2020;:1–24. [DOI] [PubMed] [Google Scholar]
- 71.Klemen ND, Wang M, Feingold PL, et al. Patterns of failure after immunotherapy with checkpoint inhibitors predict durable progression-free survival after local therapy for metastatic melanoma. 2019;:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Klemen ND, Feingold PL, Goff SL, et al. Metastasectomy Following Immunotherapy with Adoptive Cell Transfer for Patients with Advanced Melanoma. Ann Surg Oncol 2017;24(1):135–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Keung EZ, Tsai J-W, Ali AM, et al. Analysis of the immune infiltrate in undifferentiated pleomorphic sarcoma of the extremity and trunk in response to radiotherapy: Rationale for combination neoadjuvant immune checkpoint inhibition and radiotherapy. 2017;:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Roland CL, Keung EZ-Y, Lazar AJ, et al. Preliminary results of a phase II study of neoadjuvant checkpoint blockade for surgically resectable undifferentiated pleomorphic sarcoma (UPS) and dedifferentiated liposarcoma (DDLPS). JCO 2020;38(15_suppl):11505–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ariyan CE, Brady MS, Siegelbaum RH, et al. Robust Antitumor Responses Result from Local Chemotherapy and CTLA-4 Blockade. Cancer Immunol Res 2018;6(2):189–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.D’Angelo SP, Russell J, Lebbé C, et al. Efficacy and Safety of First-line Avelumab Treatment in Patients With Stage IV Metastatic Merkel Cell Carcinoma: A Preplanned Interim Analysis of a Clinical Trial. JAMA Oncol 2018;4(9):e180077–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kaufman HL, Russell J, Hamid O, et al. Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial. The Lancet Oncology 2016;17(10):1374–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.D’Angelo SP, Larkin J, Sosman JA, et al. Efficacy and Safety of Nivolumab Alone or in Combination With Ipilimumab in Patients With Mucosal Melanoma: A Pooled Analysis. JCO 2017;35(2):226–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Shoushtari AN, Munhoz RR, Kuk D, et al. The efficacy of anti-PD-1 agents in acral and mucosal melanoma. Cancer 2016;122(21):3354–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Algazi AP, Tsai KK, Shoushtari AN, et al. Clinical outcomes in metastatic uveal melanoma treated with PD-1 and PD-L1 antibodies. Cancer 2016;122(21):3344–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Heppt MV, Heinzerling L, Kähler KC, et al. Prognostic factors and outcomes in metastatic uveal melanoma treated with programmed cell death-1 or combined PD-1/cytotoxic T-lymphocyte antigen-4 inhibition. Eur J Cancer 2017;82:56–65. [DOI] [PubMed] [Google Scholar]
- 82.Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366(26):2443–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med 2017;377(14):1345–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Robert C, Schachter J, Long GV, et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med 2015;372(26):2521–32. [DOI] [PubMed] [Google Scholar]
- 85.Nghiem PT, Bhatia S, Lipson EJ, et al. PD-1 Blockade with Pembrolizumab in Advanced Merkel-Cell Carcinoma. N Engl J Med 2016;374(26):2542–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data generated for this manuscript are available from the corresponding author upon reasonable request.