Skip to main content

REVIEW article

Front. Oncol., 14 March 2023
Sec. Molecular and Cellular Oncology
This article is part of the Research Topic New Therapeutics for Soft Tissue Sarcomas View all 10 articles

New therapeutics for soft tissue sarcomas: Overview of current immunotherapy and future directions of soft tissue sarcomas

Gyuhee SeongGyuhee Seong1Sandra P. D&#x;Angelo*Sandra P. D’Angelo2*
  • 1Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, United States
  • 2Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States

Soft tissue sarcoma is a rare and aggressive disease with a 40 to 50% metastasis rate. The limited efficacy of traditional approaches with surgery, radiation, and chemotherapy has prompted research in novel immunotherapy for soft tissue sarcoma. Immune checkpoint inhibitors such as anti-CTLA-4 and PD-1 therapies in STS have demonstrated histologic-specific responses. Some combinations of immunotherapy with chemotherapy, TKI, and radiation were effective. STS is considered a ‘cold’, non-inflamed tumor. Adoptive cell therapies are actively investigated in STS to enhance immune response. Genetically modified T-cell receptor therapy targeting cancer testis antigens such as NY-ESO-1 and MAGE-A4 demonstrated durable responses, especially in synovial sarcoma. Two early HER2-CAR T-cell trials have achieved stable disease in some patients. In the future, CAR-T cell therapies will find more specific targets in STS with a reliable response. Early recognition of T-cell induced cytokine release syndrome is crucial, which can be alleviated by immunosuppression such as steroids. Further understanding of the immune subtypes and biomarkers will promote the advancement of soft tissue sarcoma treatment.

1 Introduction

Sarcomas are a rare and heterogeneous group of solid tumors of mesenchymal origin, accounting for only 1% of all adult malignancies. They can be divided broadly into soft tissue sarcomas (STS), which originate in the fat, muscle, nerve, nerve sheath, blood vessels, and other connective tissues or the bone.

More than 70 different histologic subtypes of STS have been identified (1). Soft tissue sarcoma is an aggressive disease with a 40 to 50% metastasis rate, with a 5-year survival rate of 30%. STS most commonly metastasizes to the lungs; tumors in the abdominal cavity more commonly metastasize to the liver and peritoneum (2).

The limited durable response with traditional surgery, radiation, and chemotherapy in advanced-stage sarcoma has prompted research in novel immunotherapy of soft tissue sarcoma.

1.1 Immune microenvironment of sarcoma

The tumor microenvironment (TME) comprises a tumor, stromal cells, and immune cells such as macrophages, lymphocytes, and extracellular matrix (3). Tumor cells take advantage of TME over time, and genetic/epigenetic changes of the tumor and rearrangement of TME are pivotal in tumorigenesis (4).

Tumor associated macrophages (TAMs) are distinguished components in TME. Tumors secrete high levels of colony-stimulating factor 1 (CSF-1), which converts M1 macrophage (classically activated, tumoricidal) to M2 macrophage/TAMs (alternatively activated, tumor-promoting) and stimulates tumor growth and metastasis along with CCL2 (5).

Sarcoma is traditionally considered an immunologically quiet tumor with low tumor mutational burden (1.06 mutations/Mb) and immunosuppressive TME (high levels of hypoxia-inducible factor 1 α (HIF1α), macrophages, neutrophils, and decreased T-cell levels) (6). A subset of sarcomas are sensitive to ICIs. They are ‘hot’/immune-sensitive tumors with high TMB, interferon, CD8 lymphocytes, and PD-L1 expression (7, 8).

A very recent paper highlights the significant prognostic value of systemic inflammatory indexes as a prognostic marker in terms of PFS and OS in STS patients who progressed on anthracycline. A low lymphocyte-to-monocyte ratio (LMR) was associated with worse OS (p = 0.006). Interestingly, low lymphocyte-to-monocyte ratio (LMR) was an indicator of trabectedin efficacy, which could be applied in clinical practice (9). In a previous study in 2021, 3D-cultured cells from leiomyosarcoma and undifferentiated pleomorphic sarcoma (UPS) surgical specimens were treated with trabectedin and demonstrated the involvement of ECM-associated genes such as mmps and their inhibitor timp1, emphasizing the potential role of ECM in the activity of trabectedin (10).

It was proposed that tumors with high PD-1 expression and tumor-infiltrating lymphocytes (TILs) respond well to ICIs (11). Sarcomas have relatively low PD-1 and TILs. Various studies have revealed conflicting results regarding how PD1 expression impacts prognosis. A recent review of Phase II trials demonstrated that 30% of patients with PD-L1 expression (≥1%) achieved a response. However, 7% of PD-L1 negative patients also achieved a response, underscoring the limitation of PD-L1 as a prognostic marker (12). A subsequent analysis of SARC028 revealed that higher TILs at baseline were associated with a better PFS.

In this article, we will review current immunotherapy of soft tissue sarcoma, highlighting prominent trials with immune checkpoint inhibitors and adoptive cellular therapies, including engineered T-cell receptor targeting cancer testis antigens (CTA), chimeric antigen receptor (CAR) T-cell therapies and tumor-infiltrating lymphocytes (TILs).

2 Immune checkpoint inhibitors

Immune checkpoint inhibitors (ICI) regulate critical inhibitory signals of T-cells such as PD-1/PD-L1 and CTLA-4 axes as monotherapy or in combination with chemotherapy. ICIs are FDA-approved to treat more than 50 cancer types, including advanced solid tumors, MMR-deficient tumors, and tumors with a high tumor mutation burden (13).

SARC028 was a significant Phase II trial published in 2017, which first demonstrated the efficacy of pembrolizumab (PD-1 inhibitor) in some STS, notably in undifferentiated pleomorphic sarcoma (UPS) (4 of 10) and dedifferentiated liposarcoma (dLPS) (2 of 10) (14). The final results of SARC028 expansion cohorts confirmed effectiveness in UPS, with an objective response rate (ORR) of 23%, but not in dedifferentiated/pleomorphic liposarcoma (LPS) with an ORR of 10% (15).

In the Phase II Alliance A091401 trial, patients with metastatic sarcoma were treated with nivolumab (PD-1 inhibitor) with or without ipilimumab (CTLA-4 inhibitor). Dual immune checkpoint blockade demonstrated an overall response (ORR) of 16%. Responses were confirmed in leiomyosarcoma (uterine (n=1), non-uterine (n=1)), myxofibrosarcoma (n=1), UPS (n=2), and angiosarcoma (n=1) (16). In a phase II study for advanced uterine leiomyosarcoma, none of the 12 patients responded to nivolumab alone (17). In a subsequent Phase II expansion cohort study, combination therapy of nivolumab and ipilimumab resulted in an ORR of 28.6% in UPS and 14.3% in dedifferentiated liposarcoma (18). In a DART trial by SWOG, a phase II trial of ipilimumab and nivolumab in angiosarcoma demonstrated an ORR of 25% (19). On December 2022, atezolizumab was granted FDA approval for unresectable or metastatic alveolar soft part sarcoma (ASPS) (ORR = 24%, NCT03141684).

Myxofibrosarcoma (MFS) expresses high levels of immune microenvironment markers, and some case reports support PD-1 inhibition in myxofibrosarcoma, which is further explored in a Phase II trial (ENVASARC, NCT04480502) (2023).

ICI response in soft tissue sarcoma has been modest and histologic-specific, especially in UPS, dLPS, ASPS, and angiosarcoma.

2.1 ICI and local/systemic therapy

Combinational strategies with ICI and local/systemic therapies can overcome soft tissue sarcoma resistance mechanisms. Local therapies to complement ICI consist of isolated limb infusion and radiation.

Isolated limb infusion (ILI) is a minimally invasive administration of high-dose chemotherapy to treat STS in the extremities (24). Two patients with recurrent myxofibrosarcoma responded to melphalan via ILI and pembrolizumab (1=partial response, 1=complete response) (25). This promising case prompted a subsequent Phase II trial with pembrolizumab plus the infusion of melphalan and dactinomycin (NCT04332874).

Radiation therapy is another local therapy to activate anti-tumor immunogenicity in the tumor microenvironment through the cGAS-STING pathway and subsequent CD8+ T cell activation (26, 27). There are approximately ten ongoing trials to investigate the effect of radiation in addition to ICI.

Chemotherapy enhances immunosurveillance by releasing type I interferon (IFN), and increasing M2 macrophages, CD8+ T cells, and NK cells in a tumor microenvironment (28, 29).

Two Phase II trials of doxorubicin and pembrolizumab from Pollack et al. and Livingston et al. demonstrated promising ORR of 19% in advanced sarcoma and 36.7% in advanced STS, respectively (30, 31). In a Pollack et al. study, grade 3+ treatment-related adverse effects (TRAEs) such as neutropenia (6/37), leukopenia (1/37), and febrile neutropenia (1/37), heart failure due to doxorubicin (2/37), and adrenal insufficiency (1/37) and hypothyroidism (7/37) due to pembrolizumab were observed. In a Livingston et al. study, grade 3+ TRAEs include neutropenia and leukopenia (11/30 each), and anemia (8/30). Arthralgia (3/30), fatigue (2/30), autoimmune disorder (2/30), and increased lipase (2/30) were grade 3+ TRAEs attributed to pembrolizumab. Additionally, pembrolizumab-related synovitis/myositis (n=1), autoimmune hepatitis (n=1), and autoimmune nephritis (n=1) were observed, and all patients responded to steroids. Grade 5 adverse events were not reported in both studies.

Trabectedin, in addition to ipilimumab and nivolumab, revealed an ORR of 19.5% in metastatic STS (32). Grade 4 adverse events include anemia, neutropenia, thrombocytopenia, and increased AST/ALT and CPK. Grade 5 rhabdomyolysis was observed in one patient.

Another strategy to augment immune response in STS is to combine small molecule inhibitors such as tyrosine kinase inhibitors (TKI). In the Phase II Immunosarc trial, TKI sunitinib with nivolumab in metastatic or locally advanced STS led to an ORR of 21%, with 48% of 6-month PFS (33). Wilky et al. demonstrated the efficacy of Axitinib (VEGF receptor TKI) and pembrolizumab in advanced sarcoma. None achieved a complete response. 8 out of 32 patients achieved a partial response (ORR 25.0%), with most responses occurring in ASPS (6/11, ORR 54.5%) (34).

Pembrolizumab is FDA-approved in many cancers such as advanced melanoma, Merkel Cell Carcinoma, Cutaneous Squamous Cell Carcinoma, and non-small cell lung cancer, either alone or with other therapies (3538).

Phase II trials combining systemic therapy with pembrolizumab in sarcoma are in progress: Pembrolizumab + eribulin (NCT03899805), pembrolizumab + gemcitabine (NCT03123276), pembrolizumab + lenvatinib (NCT04784247), pembrolizumab + doxorubicin (NCT03056001), pembroliumab + cabozantinib (PEMBROCABOSARC, NCT05182164), pembrolizumab + epacadostat (IDO1 Inhibitor)(NCT03414229).

Other PD-1 inhibitors in sarcoma are investigated in Phase II trials. Nivolumab + Gemcitabine/Doxorubicin/Docetaxel (GALLANT, NCT04535713), Retifanlimab (PD-1 inhibitor) + Gemcitabine/Docetaxel (NCT04577014), Sintilimab (PD-1 inhibitor) + Doxorubicin/Ifosfamide (NCT04356872) and Camrelizumab (PD-1 inhibitor) + Doxorubicin/Ifosfamide (NCT04606108) are in progress.

Future research should aim to identify biomarkers in STS to augment responses of ICI with and without local/systemic therapies in each patient.

3 Adoptive cellular therapies

Successful T-cell treatments for hematological malignancies have sparked interest in researching T-cell therapies for solid tumors such as sarcomas.

One of sarcoma’s primary immune evasion strategies is inadequate neoantigens/antigen recognition, which fails to create enough tumor-specific T cells and immune responses. Adoptive cellular therapies hope to avoid this phase by supplying a significant amount of autologous T cells specifically designed for a particular antigen. Autologous T cells are obtained from peripheral blood or the original tumor and then amplified. Potential approaches include engineered T-cell receptor (TCR) and chimeric antigen receptor (CAR) T-cell therapy and tumor-infiltrating lymphocyte (TIL) therapy with sarcoma.

3.1 Engineered T-cell receptor therapy

Cancer testis antigens (CTA) are tumor-associated antigens (TAA) that are typically present in fetal development (placenta and embryo) or at immune-privileged sites without MHC class I (testes) (39). Sarcomas express higher than normal CTAs, especially in SS and myxoid/round cell liposarcoma (40, 41). Sarcomas express a variety of CTAs such as the NY-ESO-1, MAGE, and GAGE family and fetal acetylcholine receptors (42).

NY-ESO-1 and MAGE family are intracellular antigens that must be processed and presented with MHC. TCR T cells require patients with matching HLA allele subtypes, often HLA-A2, which compose approximately 30% of the population. Modified TCR T cells recognize processed peptides via HLA-A2-specific manner and mount immune responses (43).

In 2011, Robbins et al. successfully investigated the antitumor response of NY-ESO-1-specific TCRs with high dose interleukin-2 in refractory synovial sarcoma (SS). Objective clinical responses were observed in 4 of 6 SS patients. A partial response lasted for 18 months in a patient with synovial sarcoma (44). Long-term follow-up study which enrolled 12 additional SS patients, revealed that 11 of 18 patients with SS who received anti-NY-ESO-1 TCRs responded to therapy (61%), and one had a complete response (45).

In a Phase I trial in 2018, T cells expressing NY-ESO-1c259 (Letetresgene autoleucel), a modified TCR recognizing NY-ESO-1/LAGE1a peptide, demonstrated an ORR of 50% (6/12) in metastatic SS following a lymphodepleting regimen of fludarabine and cyclophosphamide. Remarkably, self-generating pools of NY-ESO-1c259T cells persisted in vivo for at least 6 months in all patients who responded. No fatal adverse events were reported. Grade 3-4 adverse events include lymphopenia, leukopenia, neutropenia, anemia, thrombocytopenia, and hypophosphatemia. Cytokine release syndrome was reported in five patients, with median onset within 4 days and a median duration of 10 days (46).

High dose fludarabine-containing regimen is necessary for the efficacy of NY-ESO-1c259 TCR, likely correlated with elevated IL-7 and IL-15, and TAM modulation (47).

Afamitresgene autoleucel (ADP-A2M4 SPEAR TCRs directed against the MAGE-A4) revealed comparable efficacy. Phase I study with MAGE-A4c1032 TCR by Hong et al. observed an ORR of 25% in advanced solid tumors, and all partial responses were in patients with synovial sarcoma. Two patients had trial-related deaths due to aplastic anemia and CVA (48). A subsequent phase II study with afamitresgene autoleucel revealed an ORR of 40% in 25 patients with a tolerable safety profile in advanced/metastatic SS and Myxoid/Round Cell Liposarcoma (MRCLS) (49).

Although engineered TCR in advanced soft tissue sarcoma presents promising efficacy, there are some limitations to overcome, particularly the HLA-A2 requirement, manufacturing timelines/cost, and associated toxicities such as cytokine release syndrome. Furthermore, there are heterogenous CTA expressions in different types of sarcomas, and broad applicability may be limited (43).

3.2 Chimeric antigen receptor T-cell therapies

CARs are chimeric antigen receptors artificially engineered to recognize naturally occurring tumor surface antigens and activate T-cells in an MHC-independent manner (50).

C19-targeted CAR T-cell therapies for hematologic malignancies such as CD19-positive B-cell acute lymphoblastic leukemia and B-cell lymphomas have been successful. In 2022, Ciltacabtagene autoleucel, B-cell maturation antigen-directed CAR T-cell, was FDA-approved for patients with refractory or relapsed multiple myeloma who received at least four lines of therapy (CARTITUDE-1, NCT03548207). Further efforts to expand CAR T-cell therapies in solid tumors are ongoing but have not shown major significance yet.

In Phase I/II trial in HER2-positive sarcomas, including 16 osteosarcomas, one Ewing sarcoma, one primitive neuroectodermal tumor, and one desmoplastic small round cell tumor, HER2-CAR T cell therapy induced stable disease in four patients without significant toxicity (51).

In another Phase I trial, ten HER2+ refractory/metastatic patients (osteosarcoma (5), rhabdomyosarcoma (3), Ewing sarcoma (1), and synovial sarcoma (1)) were enrolled and treated with HER2-CAR T cells and lymphodepletion with either fludarabine or in combination with cyclophosphamide. At the initial follow-up at 6 weeks, 4 patients had progression, and 4 patients achieved stable disease. Overall survival at 1 year was 60% for patients treated with HER2-CAR T cells and lymphodepletion (52).

EGFR, GD2, insulin-like growth factor 1 receptor (IGF-1R), tyrosine kinase orphan-like receptor 1 (ROR1), CD44v6, and NK cell activating receptor group 2-member D (NKG2D) are potential targets in sarcoma, and early phase trials are underway to investigate the efficacy of CAR therapies for these targets.

CAR T-cell therapies will have to overcome a few obstacles in the future. CAR T-cell therapies have limited cancer-specific antigens, whereas TCRs recognize peptides presented via MHC class 1, which essentially include whole proteasome (53, 54). Until now, CAR-T therapies seek more specific targets in solid tumors, which are conserved and do not convey toxicity to healthy tissue, to improve long-term efficacy (55).

Cytokine release syndrome (CRS) is one of the adverse effects of both TCR and CAR T-cell therapy following T-cell administration. CRS is an acute, systemic response from immune stimulation in an “on-target and on-tumor” manner. T-cell therapies can also induce unexpected “on-target, off-tumor” autoimmunity, which damages healthy cells by recognizing shared antigens (5658). It is crucial to promptly recognize and treat immune-mediated adverse effects, which can be alleviated by immunosuppression such as Tocilizumab and steroids if needed.

3.3 Tumor-infiltrating lymphocytes therapies

Tumor-infiltrating lymphocytes (TIL) are extracted from tumors and administered to the patients after ex vivo expansion (59, 60). TIL had reproducible effects in melanoma. In a phase 3 trial by Rohaan et al. in 2022, TIL therapy demonstrated an ORR of 49% (41/84) in advanced melanoma (61). There has not yet demonstrated satisfactory efficacy in other solid tumors.

In 2021, Mullinax et al. investigated a rapid expansion protocol that TIL cultures from soft tissue sarcoma resection can expand enough for clinical adoptive cell therapy, which led to an ongoing Phase I trial (NCT04052334) (62).

Current challenges for TIL therapies include high cost due to the personalized nature of TIL therapies, and toxicities from high-dose IL-2, which is given post-TIL administration (63, 64).

4 Cancer vaccines

Talimogene laherparepvec (T-VEC) is an oncolytic viral immunotherapy via intratumor injection. It enhances immunogenicity via antigen presentation and tumor-specific T cells. T-VEC is the first viral immunotherapy approved for metastatic melanoma (65).

In a Phase II trial, 20 patients with advanced/metastatic sarcoma were treated with an oncolytic virus, T-VEC, with pembrolizumab, which demonstrated an ORR of 35% and a median duration of response of 56.1 weeks (66).

Vaccine therapies have been explored for decades without satisfactory results, likely due to suppressive tumor microenvironment. Current efforts are utilizing novel vectors to promote specificity and strength of immune response.

A novel study by Somaiah et al. demonstrated the efficacy of LV305, a lentivirus vector designed to induce NY-ESO-1 in dendritic cells in vivo, improving immune response against tumor cells (67). ORR was 4.2% in sarcoma (1/24 in SS).

CMB305 (a heterologous vaccine for NY-ESO-1 and TLR 4 agonist) is a good vehicle for synovial sarcoma and myxoid/round cell liposarcoma patients, and it was subsequently assessed in a Phase Ib study (68, 69). The study demonstrated a disease control rate of 61.9% and OS of 26.2 months in 64 sarcoma patients. Phase II study with CMB305 and atezolizumab (PD-L1 antibody) compared to atezolizumab alone in STS did not reveal significant improvement in PFS or OS compared to atezolizumab alone (70).

5 Future directions

Although adoptive cellular therapies offer potential individual treatments, they are still in their infancy for soft tissue sarcoma. Targeting fusion-derived cancer testis antigens such as NYESO-1 and MAGEA-4 has shown benefits in limited sarcomas such as synovial sarcoma and Myxoid/Round Cell Liposarcoma (7173).

Colony-stimulating factor-1 (CSF1) promotes “macrophage polarization”, increasing M2/M1 macrophage ratio. CSF1R inhibitor can be a potent immunomodulator by prohibiting the recruitment of TAMs into TME (74). CSF1R-targeting agents have shown a relatively tolerable safety profile but only modest clinical activity.

TTI-621 is a recombinant fusion antibody for SIRPα, a binding domain for CD47, which interrupts inhibition of macrophage phagocytosis mediated by CD47 and stimulates phagocytosis. Combination of doxorubicin with TTI-621 (anti-CD47 antibody) has shown anti-tumor effect in animal models, especially in tumors which express high number of CD47 and macrophages, such as leiomyosarcoma (75). Phase I/II study with TTI-621 alone and in combination with doxorubicin for patients with advanced leiomyosarcoma is underway (NCT04996004).

DR5 Agonist Antibody targeting the TRAIL-TNF axis, which promotes tumor-specific apoptosis, is evaluated in a Phase II study of chondrosarcomas (NCT04950075). NK cell therapies have limited data in solid tumors, and trials for sarcoma (NCT01875601, NCT02890758, NCT03420963) are currently in Phase I.

Envafolimab is a single-domain PD-L1 antibody and administered subcutaneously. There is an ongoing phase II trial evaluating envafolimab alone and with ipilimumab in undifferentiated pleomorphic sarcoma or myxofibrosarcoma (ENVASARC, NCT04480502). A multicenter phase II trial of paclitaxel alone and with nivolumab in taxane-naïve angiosarcoma patients is ongoing. (Alliance A091902, NCT04339738).

In recent years, nanotechnology has shown potential in sarcoma treatment thanks to the development of smart materials and more effective drug delivery systems. Examples include effective docetaxel-loaded mPEG-PLA nanoparticles in sarcoma-bearing mice and albumin-paclitaxel (nab-paclitaxel/Abraxane™) in osteosarcoma mice (7678). (79) Only four nano-drug delivery systems have been FDA-approved for sarcoma - Doxil (Caelyx)® for AIDS-related Kaposi’s sarcoma, DaunoXome® and Lipo-Dox® for Kaposi’s sarcoma and Liposomal mifamurtide (MEPACT) for Osteosarcoma. For locally advanced STS, there was a randomized, controlled Phase II-III trial by Bonvalot et al. in 2019 which investigated the role of NBTXR3, a radiation-enhancing nano-particle with radiotherapy compared to radiotherapy alone, demonstrated the efficacy of NBTXR3 with radiation (CR 16% vs. 8%, p = 0.044). There already exists pre-clinical evidence in 2014 which demonstrated that the chitosan nanoparticle-Methylglyoxal complex has effective antitumor properties and elicits macrophage-mediated immunity in Sarcoma-180 tumor-bearing mice (80). A Phase I trial with BO-112 (a synthetic RNA conjugated with nano-sized polyethyleneimine, which activates the immune system) with nivolumab before surgery for resectable STS, is active since 2020. (NCT04420975)

The immunosuppressive microenvironment in STS should be easier to overcome with safer and more effective next-generation immunotherapy. It is currently understood that MMR deficiency is rare and tumor mutation burden is low (3.3/Mb) in STS (7, 8184). In addition to a traditional concept of “immunologically hot” sarcoma with complex karyotypes which expresses high immune-infiltrate TME and responds well to immunotherapy, there is emerging evidence of epigenetic modulation of transcription in sarcoma, which boosts immunogenicity (85, 86). In a retrospective study of 35 patients, DNA methylation degree correlated with response to anti-PD-1 therapy in sarcoma (87).

There remains a question of whether the mutational burden or neoantigen in STS is clinically correlated to treatment response in immunotherapy. Tumor-infiltrating lymphocytes and PD-L1 expression in STS have shown conflicting prognostic significance thus far. Advancements in bioinformatics and molecular technology will guide the finding of potential biomarkers, which will help fine-tune more effective combinations for each patient in future trials.

6 Summary

Advanced soft tissue sarcoma is still a devastating diagnosis, and there are limited treatments that have long-term success rates.

This article reviewed current immunotherapy in STS, mainly immune checkpoint inhibitors alone or with additional local/systemic therapy and adoptive cell therapy, which modifies the immunogenicity of tumors and TME.

There is a dire need to identify genetic and clinical indicators of response, resistance, and toxicity in immunotherapy in STS. To better characterize histologic/molecular subtypes of STS, tissue and liquid biopsies should be more frequently utilized.

Advancement in the laboratory and clinical immunotherapy of STS for the last five years has been encouraging. By learning from each patient in clinical trials, we hope that patients with soft tissue sarcoma can benefit in the new era of immunotherapy.

Author contributions

GS writing - original draft and editing. SD conceptualization, review, and supervision. All authors contributed to the article and approved the submitted version.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. WHO Classification of Tumors Editorial Board. Soft tissue and bone tumors: WHO classification of tumours, vol. 3. 5th ed. Geneva, Switzerland: World Health Organization (2020).

Google Scholar

2. Von Mehren M, Kane JM, Agulnik M, Bui MM, Carr-Ascher J, Choy E, et al. Soft tissue sarcoma, version 2.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw (2022) 20(7):815–33. doi: 10.6004/jnccn.2022.0035

CrossRef Full Text | Google Scholar

3. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med (2018) 24(5):541–50. doi: 10.1038/s41591-018-0014-x

CrossRef Full Text | Google Scholar

4. Baghban R, Roshangar L, Jahanban-Esfahlan R, Seidi K, Ebrahimi-Kalan A, Jaymand M, et al. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal (2020) 18(1):59. doi: 10.1186/s12964-020-0530-4

CrossRef Full Text | Google Scholar

5. Scholl SM, Pallud C, Beuvon F, Hacene K, Stanley ER, Rohrschneider L, et al. Anti-colony-stimulating factor-1 antibody staining in primary breast adenocarcinomas correlates with marked inflammatory cell infiltrates and prognosis. J Natl Cancer Inst (1994) 86(2):120–6. doi: 10.1093/jnci/86.2.120

CrossRef Full Text | Google Scholar

6. Stahl D, Gentles AJ, Thiele R, Gütgemann I. Prognostic profiling of the immune cell microenvironment in ewing´s sarcoma family of tumors. Oncoimmunology (2019) 8(12):e1674113. doi: 10.1080/2162402X.2019.1674113

CrossRef Full Text | Google Scholar

7. Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med (2017) 9(1):34. doi: 10.1186/s13073-017-0424-2

CrossRef Full Text | Google Scholar

8. Keung EZ, Burgess M, Salazar R, Parra ER, Rodrigues-Canales J, Bolejack V, et al. Correlative analyses of the SARC028 trial reveal an association between sarcoma-associated immune infiltrate and response to pembrolizumab. Clin Cancer Res (2020) 26(6):1258–66. doi: 10.1158/1078-0432.CCR-19-1824

CrossRef Full Text | Google Scholar

9. Fausti V, De Vita A, Vanni S, Ghini V, Gurrieri L, Riva N, et al. Systemic inflammatory indices in second-line soft tissue sarcoma patients: Focus on Lymphocyte/Monocyte ratio and trabectedin. Cancers (2023) 15(4):1080. doi: 10.3390/cancers15041080

CrossRef Full Text | Google Scholar

10. De Vita A, Recine F, Miserocchi G, Pieri F, Spadazzi C, Cocchi C, et al. The potential role of the extracellular matrix in the activity of trabectedin in UPS and l-sarcoma: evidences from a patient-derived primary culture case series in tridimensional and zebrafish models. J Exp Clin Cancer Res (2021) 40(1):165. doi: 10.1186/s13046-021-01963-1

CrossRef Full Text | Google Scholar

11. Teng MW, Ngiow SF, Ribas A, Smyth MJ. Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res (2015) 75(11):2139–45. doi: 10.1158/0008-5472.CAN-15-0255

CrossRef Full Text | Google Scholar

12. Italiano A, Bellera C, D’Angelo S. PD1/PD-L1 targeting in advanced soft-tissue sarcomas: a pooled analysis of phase II trials. J Hematol Oncol (2020) 13(1):55. doi: 10.1186/s13045-020-00891-5

CrossRef Full Text | Google Scholar

13. Robert C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat Commun (2020) 11(1):3801. doi: 10.1038/s41467-020-17670-y

CrossRef Full Text | Google Scholar

14. Tawbi HA, Burgess M, Bolejack V, Van Tine BA, Schuetze SM, Hu J, et al. Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial. Lancet Oncol (2017) 18(11):1493–501. doi: 10.1016/S1470-2045(17)30624-1

CrossRef Full Text | Google Scholar

15. Burgess MA, Bolejack V, Schuetze S, Van Tine BA, Attia S, Riedel RF, et al. Clinical activity of pembrolizumab (P) in undifferentiated pleomorphic sarcoma (UPS) and dedifferentiated/pleomorphic liposarcoma (LPS): final results of SARC028 expansion cohorts. J Clin Oncol (2019) 37(Suppl):11015. doi: 10.1200/JCO.2019.37.15_suppl.11015

CrossRef Full Text | Google Scholar

16. D’Angelo SP, Mahoney MR, Van Tine BA, Atkins J, Milhem MM, Jahagirdar BN, et al. Nivolumab with or without ipilimumab treatment for metastatic sarcoma (Alliance A091401): two open-label, non-comparative, randomised, phase 2 trials. Lancet Oncol (2018) 19:416–26. doi: 10.1016/S1470-2045(18)30006-8

CrossRef Full Text | Google Scholar

17. Ben-Ami E, Barysauskas CM, Solomon S, Tahlil K, Malley R, Hohos M, 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: 10.1002/cncr.30738

CrossRef Full Text | Google Scholar

18. Chen JL, Mahoney MR, George S, Antonescu CR, Liebner DA, Van Tine BA, et al. A multicenter phase II study of nivolumab +/- ipilimumab for patients with metastatic sarcoma (Alliance A091401): results of expansion cohorts. J Clin Oncol (2020) 38(Suppl):11511.

Google Scholar

19. Wagner MJ, Othus M, Patel SP, Ryan C, Sangal A, Powers B, et al. Multicenter phase II trial (SWOG S1609, cohort 51) of ipilimumab and nivolumab in metastatic or unresectable angiosarcoma: a substudy of dual anti-CTLA-4 and anti-PD-1 blockade in rare tumors (DART). J Immunother Cancer (2021) 9:e002990. doi: 10.1136/jitc-2021-002990

CrossRef Full Text | Google Scholar

20. Vanni S, De Vita A, Gurrieri L, Fausti V, Miserocchi G, Spadazzi C, et al. Myxofibrosarcoma landscape: diagnostic pitfalls, clinical management and future perspectives. Ther Adv Med Oncol (2022) 14:17588359221093973. doi: 10.1177/17588359221093973

CrossRef Full Text | Google Scholar

21. Zhou M, Bui N, Lohman M, van de Rjin M, Hwang G, Ganjoo K. Long-term remission with Ipilimumab/Nivolumab in two patients with different soft tissue sarcoma subtypes and no PD-L1 expression. Case Rep Oncol (2021) 14(1):459–65. doi: 10.1159/000512828

CrossRef Full Text | Google Scholar

22. Luo Y, Min L, Zhou Y, Tang F, Lu M, Xie H, et al. Remarkable response to anti-PD1 immunotherapy in refractory metastatic high-grade myxofibrosarcoma patient: A case report. Med (Baltimore) (2021) 100(12):e25262. doi: 10.1097/MD.0000000000025262

CrossRef Full Text | Google Scholar

23. Lambden JP, Kelsten MF, Schulte BC, Abbinanti S, Hayes JP, Villaflor V, et al. Metastatic myxofibrosarcoma with durable response to temozolomide followed by atezolizumab: A case report. Oncologist (2021) 26(7):549–53. doi: 10.1002/onco.13728

CrossRef Full Text | Google Scholar

24. Creech O Jr, Krementz ET, Ryan RF, Winblad JN. Chemotherapy of cancer: regional perfusion utilizing an extracorporeal circuit. Ann Surg (1958) 148:616–32. doi: 10.1097/00000658-195810000-00009

CrossRef Full Text | Google Scholar

25. Bartlett EK, D'Angelo SP, Kelly CM, Siegelbaum RH, Fisher C, Antonescu CR, et al. Case report: Response to regional melphalan via limb infusion and systemic PD1 blockade in recurrent myxofibrosarcoma: A report of 2 cases. Front Oncol (2021) 11:725484. doi: 10.3389/fonc.2021.725484

CrossRef Full Text | Google Scholar

26. Constanzo J, Faget J, Ursino C, Badie C, Pouget JP. Radiation-induced immunity and toxicities: The versatility of the cGAS-STING pathway. Front Immunol (2021) 12:680503. doi: 10.3389/fimmu.2021.680503

CrossRef Full Text | Google Scholar

27. Jiang M, Chen P, Wang L, Li W, Chen B, Liu Y, et al. cGAS-STING, an important pathway in cancer immunotherapy. J Hematol Oncol (2020) 13(1):81. doi: 10.1186/s13045-020-00916-z

CrossRef Full Text | Google Scholar

28. Wang YJ, Fletcher R, Yu J, Zhang L. Immunogenic effects of chemotherapy-induced tumor cell death. Genes Dis (2018) 5(3):194–203. doi: 10.1016/j.gendis.2018.05.003

CrossRef Full Text | Google Scholar

29. Fridlender ZG, Sun J, Singhal S, Kapoor V, Cheng G, Suzuki E, et al. Chemotherapy delivered after viral immunogene therapy augments antitumor efficacy via multiple immune-mediated mechanisms. Mol Ther (2010) 18(11):1947–59. doi: 10.1038/mt.2010.159

CrossRef Full Text | Google Scholar

30. Pollack SM, Redman MW, Baker KK, Wagner MJ, Schroeder BA, Loggers ET, et al. Assessment of doxorubicin and pembrolizumab in patients with advanced anthracycline-naive sarcoma: a phase 1/2 nonrandomized clinical trial. JAMA Oncol (2020) 6:1778–82. doi: 10.1001/jamaoncol.2020.3689

CrossRef Full Text | Google Scholar

31. Livingston MB, Jagosky MH, Robinson MM, Ahrens WA, Benbow JH, Farhangfar CJ, et al. Phase II study of pembrolizumab in combination with doxorubicin in metastatic and unresectable soft-tissue sarcoma. Clin Cancer Res (2021) 27:6424–31. doi: 10.1158/1078-0432.CCR-21-2001

CrossRef Full Text | Google Scholar

32. Gordon EM, Chua-Alcala VS, Kim K, Dy PS, Paz MK, Angel N, et al. SAINT: results of an expanded phase II study using safe amounts of ipilimumab (I), nivolumab (N), and trabectedin (T) as first-line treatment of advanced soft tissue sarcoma [NCT03138161]. J Clin Oncol (2020) 38(Suppl):11520. doi: 10.1200/JCO.2020.38.15_suppl.11520

CrossRef Full Text | Google Scholar

33. Martin-Broto J, Hindi N, Grignani G, Martinez-Trufero J, Redondo A, Valverde C, et al. Nivolumab and sunitinib combination in advanced soft tissue sarcomas: a multicenter, single-arm, phase Ib/II trial. J Immunother Cancer (2020) 8:e001561. doi: 10.1136/jitc-2020-001561

CrossRef Full Text | Google Scholar

34. Wilky BA, Trucco MM, Subhawong TK, Florou V, Park W, Kwon D, et al. Axitinib plus pembrolizumab in patients with advanced sarcomas including alveolar soft part sarcoma: a single-arm,phase 2 trial. Lancet Oncol (2019) 20(6):837–48. doi: 10.1016/S1470-2045(19)30153-6

CrossRef Full Text | Google Scholar

35. Luke JJ, Rutkowski P, Queirolo P, Del Vecchio M, Mackiewicz J, Chiarion-Sileni V, et al. Pembrolizumab versus placebo as adjuvant therapy in completely resected stage IIB or IIC melanoma (KEYNOTE-716): a randomised, double-blind, phase 3 trial. Lancet (2022) 399(10336):1718–29. doi: 10.1016/S0140-6736(22)00562-1

CrossRef Full Text | Google Scholar

36. Nghiem P, Bhatia S, Lipson EJ, Sharfman WH, Kudchadkar RR, Brohl AS, et al. Durable tumor regression and overall survival in patients with advanced merkel cell carcinoma receiving pembrolizumab as first-line therapy. J Clin Oncol (2019) 37(9):693–702. doi: 10.1200/JCO.18.01896

CrossRef Full Text | Google Scholar

37. Grob JJ, Gonzalez R, Basset-Seguin N, Vornicova O, Schachter J, Joshi A, et al. Pembrolizumab monotherapy for recurrent or metastatic cutaneous squamous cell carcinoma: A single-arm phase II trial (KEYNOTE-629). J Clin Oncol (2020) 38(25):2916–25. doi: 10.1200/JCO.19.03054

CrossRef Full Text | Google Scholar

38. Mok TSK, Wu YL, Kudaba I, Kowalski DM, Cho BC, Turna HZ, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet (2019) 393(10183):1819–30. doi: 10.1016/S0140-6736(18)32409-7

CrossRef Full Text | Google Scholar

39. Fratta E, Coral S, Covre A, Parisi G, Colizzi F, Danielli R, et al. The biology of cancer testis antigens: putative function, regulation and therapeutic potential. Mol Oncol (2011) 5(2):164–82. doi: 10.1016/j.molonc.2011.02.001

CrossRef Full Text | Google Scholar

40. Jungbluth AA, Antonescu CR, Busam KJ, Iversen K, Kolb D, Coplan K, et al. Monophasic and biphasic synovial sarcomas abundantly express cancer/testis antigen NY-ESO-1 but not MAGE-A1 or CT7. Int J Cancer (2001) 94(2):252–6. doi: 10.1002/ijc.1451

CrossRef Full Text | Google Scholar

41. Hemminger JA, Ewart Toland A, Scharschmidt TJ, Mayerson JL, Kraybill WG, Guttridge DC, et al. The cancer-testis antigen NY-ESO-1 is highly expressed in myxoid and round cell subset of liposarcomas. Mod Pathol (2013) 26(2):282–8. doi: 10.1038/modpathol.2012.133

CrossRef Full Text | Google Scholar

42. Mata M, Gottschalk S. Adoptive cell therapy for sarcoma. Immunotherapy (2015) 7(1):21–35. doi: 10.2217/imt.14.98

CrossRef Full Text | Google Scholar

43. Garrido F, Aptsiauri N, Doorduijn EM, Garcia Lora AM, van Hall T. The urgent need to recover MHC class I in cancers for effective immunotherapy. Curr Opin Immunol (2016) 39:44–51. doi: 10.1016/j.coi.2015.12.007

CrossRef Full Text | Google Scholar

44. Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol (2011) 29:917–24. doi: 10.1200/JCO.2010.32.2537

CrossRef Full Text | Google Scholar

45. Robbins PF, Kassim SH, Tran TLN, Crystal JS, Morgan RA, Feldman SA, 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. Clin Cancer Res (2015) 21:1019–27. doi: 10.1158/1078-0432.CCR-14-2708

CrossRef Full Text | Google Scholar

46. D’Angelo SP, Melchiori L, Merchant MS, Bernstein D, Glod J, Kaplan R, et al. Antitumor activity associated with prolonged persistence of adoptively transferred NY-ESO-1 c259T cells in synovial sarcoma. Cancer Discovery (2018) 8:944–57. doi: 10.1158/2159-8290.CD-17-1417

CrossRef Full Text | Google Scholar

47. Ramachandran I, Lowther DE, Dryer-Minnerly R, Wang R, Fayngerts S, Nunez D, et al. Systemic and local immunity following adoptive transfer of NY-ESO-1 SPEAR T cells in synovial sarcoma. J Immunother Cancer (2019) 7:276. doi: 10.1186/s40425-019-0762-2

CrossRef Full Text | Google Scholar

48. Hong DS, Van Tine BA, Olszanski AJ, Johnson ML, Liebner DA, Trivedi T, et al. Phase I dose escalation and expansion trial to assess the safety and efficacy of ADP-A2M4 SPEAR T cells in advanced solid tumors. J Clin Oncol (2020) 38(Suppl):102. doi: 10.1200/JCO.2020.38.15_suppl.102

CrossRef Full Text | Google Scholar

49. D’Angelo SP, Van Tine BA, Attia S, Blay JY, Strauss SJ, Valverde Morales CM, et al. SPEARHEAD-1: a phase 2 trial of afamitresgene autoleucel (formerly ADP-A2M4) in patients with advanced synovial sarcoma or myxoid/round cell liposarcoma. J Clin Oncol (2021) 39(Suppl):11504.

Google Scholar

50. Poorebrahim M, Mohammadkhani N, Mahmoudi R, Gholizadeh M, Fakhr E, Cid-Arregui A. TCR-like CARs and TCR-CARs targeting neoepitopes: an emerging potential. Cancer Gene Ther (2021) 28(6):581–9. doi: 10.1038/s41417-021-00307-7

CrossRef Full Text | Google Scholar

51. Ahmed N, Brawley VS, Hegde M, Robertson C, Ghazi A, Gerken C, 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: 10.1200/JCO.2014.58.0225

CrossRef Full Text | Google Scholar

52. Navai SA, Derenzo C, Joseph S, Sanber K, Byrd TT, Zhang H, 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. doi: 10.1158/1538-7445.AM2019-LB-147

CrossRef Full Text | Google Scholar

53. Walseng E, Köksal H, Sektioglu IM, Fåne A, Skorstad G, Kvalheim G, et al. A TCR-based chimeric antigen receptor. Sci Rep (2017) 7:1–10. doi: 10.1038/s41598-017-11126-y

CrossRef Full Text | Google Scholar

54. Akatsuka Y. TCR-like CAR-T cells targeting MHC-bound minor histocompatibility antigens. Front Immunol (2020) 11:257. doi: 10.3389/fimmu.2020.00257

CrossRef Full Text | Google Scholar

55. Knochelmann HM, Smith AS, Dwyer CJ, Wyatt MM, Mehrotra S, Paulos CM, et al. CAR T cells in solid tumors: Blueprints for building effective therapies. Front Immunol (2018) 9:1740. doi: 10.3389/fimmu.2018.01740

CrossRef Full Text | Google Scholar

56. Yang JC. Toxicities associated with adoptive T-cell transfer for cancer. Cancer J (2015) 21(6):506–9. doi: 10.1097/PPO.0000000000000157

CrossRef Full Text | Google Scholar

57. Sun S, Hao H, Yang G, Zhang Y, Fu Y. Immunotherapy with CAR-modified T cells: Toxicities and overcoming strategies. J Immunol Res (2018) 2018:2386187. doi: 10.1155/2018/2386187

CrossRef Full Text | Google Scholar

58. Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood (2016) 127(26):3321–30. doi: 10.1182/blood-2016-04-703751

CrossRef Full Text | Google Scholar

59. Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res (2011) 17(13):4550–7. doi: 10.1158/1078-0432.CCR-11-0116

CrossRef Full Text | Google Scholar

60. Andersen R, Donia M, Ellebaek E, Borch TH, Kongsted P, Iversen TZ, et al. Long-lasting complete responses in patients with metastatic melanoma after adoptive cell therapy with tumor- infiltrating lymphocytes and an attenuated IL2 regimen. Clin Cancer Res (2016) 22(15):3734–45. doi: 10.1158/1078-0432.CCR-15-1879

CrossRef Full Text | Google Scholar

61. Rohaan MW, Borch TH, van den Berg JH, Met Ö, Kessels R, Geukes Foppen MH, et al. Tumor-infiltrating lymphocyte therapy or ipilimumab in advanced melanoma. N Engl J Med (2022) 387(23):2113–25. doi: 10.1056/NEJMoa2210233

CrossRef Full Text | Google Scholar

62. Mullinax JE, Hall M, Beatty M, Weber AM, Sannasardo Z, Svrdlin T, et al. Expanded tumor-infiltrating lymphocytes from soft tissue sarcoma have tumor-specific function. J Immunother (2021) 44(2):63–70. doi: 10.1097/CJI.0000000000000355

CrossRef Full Text | Google Scholar

63. Retel VP, Steuten LM, Mewes JC, van Harten WH. Early cost-effectiveness modeling for tumor infiltrating lymphocytes (TIL) -treatment versus ipilimumab in metastatic melanoma patients. Value Health (2014) 17(7):A640. doi: 10.1016/j.jval.2014.08.2307

CrossRef Full Text | Google Scholar

64. Nguyen LT, Saibil SD, Sotov V, Le MX, Khoja L, Ghazarian D, et al. Phase II clinical trial of adoptive cell therapy for patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and low-dose interleukin-2. Cancer Immunol Immunother (2019) 68:773–85. doi: 10.1007/s00262-019-02307-x

CrossRef Full Text | Google Scholar

65. Ferrucci PF, Pala L, Conforti F, Cocorocchio E. Talimogene laherparepvec (T-VEC): An intralesional cancer immunotherapy for advanced melanoma. Cancers (Basel) (2021) 13(6):1383. doi: 10.3390/cancers13061383

CrossRef Full Text | Google Scholar

66. Kelly CM, Antonescu CR, Bowler T, Munhoz R, Chi P, Dickson MA, et al. Objective response rate among patients with locally advanced or metastatic sarcoma treated with talimogene laherparepvec in combination with pembrolizumab. JAMA Oncol (2020) 6:402–8. doi: 10.1001/jamaoncol.2019.6152

CrossRef Full Text | Google Scholar

67. Somaiah N, Block MS, Kim JW, Shapiro GI, Do KT, Hwu P, et al. First-in-class, first-in-human study evaluating LV305, a dendritic-cell tropic lentiviral vector, in sarcoma and other solid tumors expressing NY-ESO-1. Clin Cancer Res (2019) 25:58085817. doi: 10.1158/1078-0432.CCR-19-1025

CrossRef Full Text | Google Scholar

68. Pollack SM. The potential of the CMB305 vaccine regimen to target NY-ESO-1 and improve outcomes for synovial sarcoma and myxoid/round cell liposarcoma patients. Expert Rev Vaccin (2018) 17(2):107–14. doi: 10.1080/14760584.2018.1419068

CrossRef Full Text | Google Scholar

69. Somaiah N, Chawla SP, Block MS, Morris JC, Do K, Kim JW, et al. A phase 1b study evaluating the safety, tolerability, and immunogenicity of CMB305, a lentiviral-based prime-boost vaccine regimen, in patients with locally advanced, relapsed, or metastatic cancer expressing NY-ESO-1. Oncoimmunology (2020) 9(1):1847846. doi: 10.1080/2162402X.2020.1847846

CrossRef Full Text | Google Scholar

70. Chawla SP, Van Tine BA, Pollack SM, Ganjoo KN, Elias AD, Riedel RF, et al. Phase II randomized study of CMB305 and atezolizumab compared with atezolizumab alone in soft-tissue sarcomas expressing NY-ESO-1. J Clin Oncol (2021) 40(12):1291–1300 doi: 10.1200/JCO.20.03452.

CrossRef Full Text | Google Scholar

71. Matsuzaki A, Suminoe A, Hattori H, Hoshina T, Hara T. Immunotherapy with autologous dendritic cells and tumor-specific synthetic peptides for synovial sarcoma. J Pediatr Hematol Oncol (2002) 24:220–3. doi: 10.1097/00043426-200203000-00012

CrossRef Full Text | Google Scholar

72. Dagher R, Long LM, Read EJ, Leitman SF, Carter CS, Tsokos M, et al. Pilot trial of tumor-specific peptide vaccination and continuous infusion interleukin-2 in patients with recurrent Ewing sarcoma and alveolar rhabdomyosarcoma: an inter-institute NIH study. Med Pediatr Oncol (2002) 38:158–64. doi: 10.1002/mpo.1303

CrossRef Full Text | Google Scholar

73. Kawaguchi S, Tsukahara T, Ida K, Kimura S, Murase M, Kano M, et al. SYT-SSX breakpoint peptide vaccines in patients with synovial sarcoma: a study from the Japanese musculoskeletal oncology group. Cancer Sci (2012) 103:1625–30. doi: 10.1111/j.1349-7006.2012.02370.x

CrossRef Full Text | Google Scholar

74. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol (2002) 23(11):549–55. doi: 10.1016/s1471-4906(02)02302-5

CrossRef Full Text | Google Scholar

75. Chawla SP, Kelly CM, Gordon EM, Quon DV, Moradkhani A, Chua-Alcala VS, et al. TTI-621-03: A phase I/II study of TTI-621 in combination with doxorubicin in patients with unresectable or metastatic high-grade leiomyosarcoma (LMS). J Clin Oncol (2022) 40:16_suppl:TPS11593–TPS11593. doi: 10.1200/JCO.2022.40.16_suppl.TPS11593

CrossRef Full Text | Google Scholar

76. Mercatali L, Vanni S, Miserocchi G, Liverani C, Spadazzi C, Cocchi C, et al. The emerging role of cancer nanotechnology in the panorama of sarcoma. Front Bioeng Biotechnol (2022) 10:953555. doi: 10.3389/fbioe.2022.953555

CrossRef Full Text | Google Scholar

77. Chen J, Ning E, Wang Z, Jing Z, Wei G, Wang X, et al. Docetaxel loaded mPEG-PLA nanoparticles for sarcoma therapy: preparation, characterization, pharmacokinetics, and anti-tumor efficacy. Drug Deliv (2021) 28(1):1389–96. doi: 10.1080/10717544.2021.1945167

CrossRef Full Text | Google Scholar

78. Yang Y, Niu X, Zhang Q, Hao L, Ding Y, Xu H. The efficacy of abraxane on osteosarcoma xenografts in nude mice and expression of secreted protein, acidic and rich in cysteine. Am J Med Sci (2012) 344(3):199–205. doi: 10.1097/MAJ.0b013e31823e62e5

CrossRef Full Text | Google Scholar

79. Wagner LM, Yin H, Eaves D, Currier M, Cripe TP. Preclinical evaluation of nanoparticle albumin-bound paclitaxel for treatment of pediatric bone sarcoma. Pediatr Blood Cancer (2014) 61(11):2096–8. doi: 10.1002/pbc.25062

CrossRef Full Text | Google Scholar

80. Chakrabarti A, Talukdar D, Pal A, Ray M. Immunomodulation of macrophages by methylglyoxal conjugated with chitosan nanoparticles against sarcoma-180 tumor in mice [published correction appears in cell immunol. 2014 mar-Apr;288(1-2):66]. Cell Immunol (2014) 287(1):27–35. doi: 10.1016/j.cellimm.2013.11.006

CrossRef Full Text | Google Scholar

81. Cancer Genome Atlas Research NetworkComprehensive and integrated genomic characterization of adult soft tissue sarcomas. Cell (2017) 171(4):950.e28.e28. doi: 10.1016/j.cell.2017.10.014

CrossRef Full Text | Google Scholar

82. Campbell BB, Light N, Fabrizio D, Zatzman M, Fuligni F, de Borja R, et al. Comprehensive analysis of hypermutation in human cancer. Cell (2017) 171(5):1042.e10.e10. doi: 10.1016/j.cell.2017.09.048

CrossRef Full Text | Google Scholar

83. Doyle LA, Nowak JA, Nathenson MJ, Thornton K, Wagner AJ, Johnson JM, et al. Characteristics of mismatch repair deficiency in sarcomas. Mod Pathol (2019) 32(7):977–87. doi: 10.1038/s41379-019-0202-3

CrossRef Full Text | Google Scholar

84. Campanella NC, Penna V, Ribeiro G, Abrahão-Machado LF, Scapulatempo-Neto C, Reis RM, et al. Absence of microsatellite instability in soft tissue sarcomas. Pathobiology (2015) 82(1):36–42. doi: 10.1159/000369906

CrossRef Full Text | Google Scholar

85. Keenan TE, Burke KP, Van Allen EM. Genomic correlates of response to immune checkpoint blockade. Nat Med (2019) 25(3):389–402. doi: 10.1038/s41591-019-0382-x

CrossRef Full Text | Google Scholar

86. Tazzari M, Bergamaschi L, De Vita A, Collini P, Barisella M, Bertolotti A, et al. Molecular determinants of soft tissue sarcoma immunity: Targets for immune intervention. Int J Mol Sci (2021) 22(14):7518. doi: 10.3390/ijms22147518

CrossRef Full Text | Google Scholar

87. Starzer AM, Berghoff AS, Hamacher R, Tomasich E, Feldmann K, Hatziioannou T, et al. Tumor DNA methylation profiles correlate with response to anti-PD-1 immune checkpoint inhibitor monotherapy in sarcoma patients. J Immunother Cancer (2021) 9(3):e001458. doi: 10.1136/jitc-2020-001458

CrossRef Full Text | Google Scholar

Keywords: soft tissue sarcoma, immune checkpoint inhibitor, adoptive immunotherapy, cancer testis antigen, T-cell receptor therapy, chimeric antigen receptor (CAR) T-cell, tumor-infiltrating lymphocyte, tumor microenvironment

Citation: Seong G and D’Angelo SP (2023) New therapeutics for soft tissue sarcomas: Overview of current immunotherapy and future directions of soft tissue sarcomas. Front. Oncol. 13:1150765. doi: 10.3389/fonc.2023.1150765

Received: 24 January 2023; Accepted: 27 February 2023;
Published: 14 March 2023.

Edited by:

Marla Weetall, PTC Therapeutics (United States), United States

Reviewed by:

Jessica Crystal, University of Miami, United States
Alessandro De Vita, Scientific Institute of Romagna for the Study and Treatment of Tumors (IRCCS), Italy

Copyright © 2023 Seong and D’Angelo. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Sandra P. D’Angelo, dangelos@mskcc.org

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.