Abstract
Children and adolescents with high-risk (metastatic and relapsed) solid tumors have poor outcomes despite intensive multimodal therapy, and there is a pressing need for novel therapeutic strategies. Adoptive cellular therapy (ACT) has demonstrated activity in multiple adult cancer types, and opportunity exists to expand the use of this therapy in children. Employment of immunotherapy in the pediatric population has realized only modest overall clinical trial results, with success thus far restricted mainly to antibody-based therapies and chimeric antigen receptor T-cell therapies for lymphoid malignancy. As we improve our understanding of the orchestrated cellular and molecular mechanisms involved in ACT, this will provide biologic insight and improved ACT strategies for pediatric malignancies. This review focuses on ACT strategies outside of chimeric antigen receptor T-cell therapy, including completed and ongoing clinical trials, and highlights promising preclinical data in tumor-infiltrating lymphocytes that enhance the clinical efficacy of ACT for high-risk pediatric solid tumors.
Keywords: adoptive cellular therapy, pediatric, solid tumors
Solid tumors account for approximately 25% of all pediatric cancers, when excluding tumors of the central nervous system.1 The most common types of pediatric malignant solid tumors (pMST) include neuroblastoma, sarcomas, and Wilms tumor.2,3 Some subsets of pMST (e.g., favorable histology Wilms tumor and germ cell tumors) demonstrate chemotherapy sensitivity and high curative potential. However, most patients with high-risk pMST (defined here as metastatic and relapsed pMST) have unsatisfactory outcomes despite combinatorial intervention with chemotherapy, surgical resection, and/or radiation that carry high treatment-related toxicity.4-9
Progress in immunotherapeutic approaches to cancer treatment, including augmenting or even replacing cytotoxic chemotherapy, continues to accelerate in both adult and pediatric malignancies. Despite these advancements, most pediatric cancers including pMST do not yet incorporate immunotherapy in standard-of-care practices. The purpose of this review is to highlight successes and challenges in immunotherapy for pediatric cancer with a focus on pMST, as well as present new opportunities for adoptive cellular therapy (ACT) outside of chimeric antigen receptor T-cell (CAR-T) therapy in this patient population.
IMMUNOTHERAPY SUCCESSES AND CHALLENGES IN pMST
Monoclonal antibody therapy is the most widely used immunotherapy in pediatric oncology to date. Their successes in pediatric leukemia and lymphoma were thoroughly reviewed in a recent publication.10 In regard to pMST, extensive investigation of antibody therapy targeting disialoganglioside (GD2) expressed by neuroectodermal cancers has improved survival rates and become a frontline immunotherapy in high-risk neuroblastoma.11-13 Thorough reviews of anti-GD2 in neuroblastoma have been published.14 Briefly, treatment consists of administration of a GD2-specific monoclonal antibody (dinutuximab, naxitamab, etc.), often in combination with isotretinoin, interleukin 2 (IL-2) and/or granulocyte-macrophage colony-stimulating factor.12,15 Unfortunately, use of GD2-specific antibodies in other pMST such as osteosarcoma and soft tissue sarcoma has shown limited responses accompanied with multiple toxicities including severe leg pain, vision dysfunction, motor weakness, and loss of vibratory sensation.13,16-18 One death, possibly attributable to dinutuximab given on a 2-day schedule plus cytokine therapy, was reported on a phase II study in relapsed osteosarcoma.19 The effectiveness of this therapy relies on host immune-cell–mediated antibody-dependent cellular cytotoxicity (ADCC), especially natural killer (NK) cells. One factor that may influence the potency of ADCC and clinical outcomes is genetic polymorphisms of the Fc-γ-receptor expressed on the immune cells and killer cell immunoglobulin-like receptor (KIR) and KIR ligand expressed by cancer cells. One cohort of neuroblastoma patients treated with anti-GD2 therapy showed those with high-affinity Fc-γ receptor and the activating KIR 2DS2 had improved event-free survival and increased ADCC.20
Immune checkpoint inhibition (ICI), including monoclonal antibody therapy targeting programmed cell death protein 1 (PD1), programmed death-ligand 1, and cytotoxic T-lymphocyte–associated protein 4, is now widely used in many adult solid tumors. One factor strongly correlating with patient response to ICI is tumor mutational burden (TMB; the sum of mutations in a tumor), which correlates with the presence of neoantigens.21 Tumors with high TMB have increased prevalence of suppressed tumor-specific T cells, leading to higher potential for response to ICI.22 Unfortunately, the TMB in pMST is generally quite low, and predicted neoantigens are present at low levels relative to common adult malignancies such as cutaneous melanoma and non–small cell lung cancer.5,23,24 With the lack of neoantigens, minimal tumor-specific T-cell activation has led to largely disappointing clinical trials of single-agent ICI in pMST, including Children's Oncology Group phase I/II trial ADVL1412 with nivolumab alone and in combination with ipilimumab, KEYNOTE-051 with pembrolizumab, and adult trials that extended down to childhood ages (e.g., SARC028 with pembrolizumab in bone sarcomas, NCT03141684 with atezolizumab in alveolar soft-part sarcoma.25-28 With the exception of alveolar soft-part sarcoma, very few or no objective responses were seen in pediatric patients with solid tumors. Leading groups such as ACCELERATE have suggested that ICI agents be studied in combination with other immunotherapies in pediatric histologies.29 Some combination strategies are already in clinical trials for pMST, including combining anti-PD1 with the DNA methylation inhibitor 5-azacitidine for relapsed osteosarcoma (NCT03628209), a neuroblastoma cancer vaccine plus anti-PD1 (NCT04239040), the multityrosine kinase inhibitor regorafenib with anti-PD1 for osteosarcoma (SARC038 - NCT04803877), and the multityrosine kinase inhibitor sunitinib with anti-PD1 and standard chemotherapy for bone and soft tissue sarcomas (NCT03277924).
ACT APPROACHES IN pMST
Adoptive cell therapies come in 2 main varieties: those that redirect immune cell specificity (e.g., CAR T cells and T-cell receptor [TCR]–engineered T cells, CAR NK cells) and those that seek to augment the function of preexisting endogenous T-cell responses by ex vivo manipulation or boosting after adoptive transfer. We will first discuss advances in therapies that seek to redirect immune cell specificity. Chimeric antigen receptor T-cell therapy represents an ACT strategy with clinical success in hematologic malignancies, including B-cell acute lymphoblastic leukemia, chronic lymphocytic leukemia, and large B-cell lymphoma.30-32 Chimeric antigen receptor T-cell therapy involves collection and expansion of circulating lymphocytes via apheresis with subsequent engineering of T cells via viral transduction to express a CAR that enables targeting of a specific tumor-associated antigen (TAA). Because tumor cells often downregulate their human leukocyte antigen (HLA) as an immune escape mechanism, this engineered T-cell approach is a valuable strategy as it bypasses the need for tumor antigen cross-presentation on HLA to T cells and instead enables direct tumor recognition by CAR-redirected T cells.33 The technique also allows the generation of large numbers of T cells with identical antigen specificity mixing both CD4+ and CD8+ T cells. Chimeric antigen receptor T-cell therapy does carry risk of severe toxicities including cytokine-release syndrome and neurotoxicity.34 Most CAR-T clinical trial successes in pediatrics have applied to lymphoid malignancy, specifically B-cell acute lymphoblastic leukemia.35
Chimeric antigen receptor T-cell therapy trials conducted in pMST, including targeting of GD2 and human epidermal growth factor receptor 2, have thus far demonstrated limited efficacy.36,37 A key obstacle to expanded utilization of CAR-T in pMST involves the identification of candidate TAAs, as most pediatric cancer types have limited known TAAs. Even with identification of novel TAAs, the heterogeneity of expression within a single tumor and potential for antigen loss and immune escape may dramatically limit the efficacy of single-antigen targeted CAR T-cell therapy.
In regard to TCR-engineered T cells, groups have cloned the α and β chains of the TCR, which can then be expressed by genetic engineering of peripheral T cells to enable infusion of large numbers of tumor-specific T cells that target the antigen presented in a major histocompatibility complex-dependent setting setting.7 Genetically modified TCR therapy has been tested in synovial sarcoma (SS).38,39 Seventy percent of SSs express the cancer testis antigen NY-ESO-1, which is considered an ideal target because of its immunogenicity and lack of expression in nonmalignant tissues.40 Unfortunately, objective clinical responses have been hampered by insufficient induction of immune response and tumor escape from immune recognition; thus, more recent efforts have focused on overcoming these barriers.40 A pilot trial testing genetically modified autologous T cells targeting NY-ESO-1 in advanced sarcoma or melanoma patients was conducted in combination with concurrent dendritic cell (DC) vaccination with or without cytotoxic T-lymphocyte–associated protein 4 targeting antibody, ipilimumab.41 Although this trial included only patients older than 16 years, the diagnoses enrolled have overlap with the pediatric population. Of 6 sarcoma patients receiving ACT with DC vaccination (3 SSs, 1 osteosarcoma, 1 liposarcoma, and 1 malignant peripheral nerve sheath tumor), 4 had transient tumor regression on PET/CT. Two SS patients achieved a partial response (PR) on day 90 of the study. Rapid expansion of NY-ESO-1–specific T cells in peripheral blood was evidenced by cell levels peaking within 2 weeks of being infused. Tumor infiltration of NY-ESO-1–specific T cells was demonstrated using sequencing of tumor biopsies, and T cells were shown over time to shift away from a memory phenotype toward terminally differentiated effector T cells. Results demonstrated that engineered T cells expressing transgenic TCR infused in conjunction with pulsed DC is both feasible and capable of providing some antitumor activity.41
Other trials attempting to enhance the clinical activity of NY-ESO-1–targeting T cells have been reported.38,42 Using autologous T cells whose TCRs were engineered for increased affinity for HLA-A2–restricted NY-ESO-1/LAGE1a–derived peptide, investigators examined safety and efficacy in targeting metastatic SS. Among 12 patients, 6 (50%) had objective response of median duration 30.9 weeks. Engineered T cells persisted in peripheral blood for 6 months in the responsive patients. Following ex vivo expansion, most T cells were characterized as having effector memory phenotype, whereas most engineered T cells that persisted in responders were characterized as central memory and stem-cell memory subsets, sustaining polyfunctionality and evading exhaustion. Results indicated the engineered T cells underwent prolonged regeneration to provide multiclonal, antigen-specific effector cells, affording a sustained antitumor response.38 Another trial utilizing T cells with affinity-enhanced TCR to target advanced-stage SS expressing NY-ESO-1–specific HLA-A*02–restricted peptide SLLMWITQC (NY-ESO-1 SPEAR T cells) included 42 patients.42 Efficacy was demonstrated with 15 objective responses (1 with complete response [CR], 14 with PR). Abrupt expansion of engineered T-cell postinfusion correlated with high tumor expression of NY-ESO-1.42 This approach is being further expanded tweaking the NY-ESO-1–targeted TCR-engineered T cells either to express CD8-α to improve stabilization of the TCR signaling or to express a dominant negative transforming growth factor β (TGF-β) receptor to reduce the suppressive signals from TGF-β in the tumor microenvironment TME; NCT03967223).
PREVIOUS NON–CAR-T ACT TRIALS IN pMST
The rest of this review will focus on use of ACT approaches that do not redirect immune cell specificity, with special focus on the therapeutic potential of tumor-infiltrating lymphocyte (TIL) ACT. Most non–CAR-T ACT trials involving pMST have been combined pediatric/adult trials or trials for adolescents and young adults with peripheral blood serving as the immune cell source. In an attempt to address the poor prognosis of metastatic Ewing sarcoma (ES) and metastatic alveolar rhabdomyosarcoma, a clinical trial aimed at consolidating remission in 52 metastatic and recurrent patients was undertaken in children and young adults.43 Before chemotherapy, blood was collected with apheresis for isolation of immune cells. After standard multimodal therapy, 30 patients continued on the trial and were divided into 3 cohorts. All cohorts received autologous T cells in conjunction with a DC vaccine pulsed with ES or alveolar rhabdomyosarcoma translocation breakpoint peptides and a control E7 peptide from human papillomavirus. Interleukin 2 was administered in moderate-dose, low-dose, or none to cohorts 1, 2, and 3, respectively. All patients apheresed had a 31% 5-year survival, whereas those receiving ACT had a 43% 5-year survival. Treatment was well tolerated with minimal toxicity, and patients analyzed had favorable influenza immune responses implying functional immune capacity following their up-front cytotoxic chemotherapy treatment. Cohort 1 (moderate-dose IL-2) seemed to have improved immune responses. Investigators concluded that despite the profound lymphopenia and alteration of T-cell subsets known to occur with sarcoma chemotherapy, this did not preclude use of postchemotherapy ACT.43
Patients with recurrent or refractory Epstein-Barr virus (EBV)–positive nasopharyngeal carcinoma have been studied in a phase I/II trial enrolling children and adults with autologous, EBV-specific cytotoxic T lymphocytes (EBV-CTLs).44 This approach utilizes known TAAs (EBVantigens) with ACT attempting to infuse large numbers of tumor-specific T cells expanded and activated ex vivo and outside the tumor environment. Of 23 total patients, 8 patients were in remission, and 15 had active disease at the time of cell infusion. No significant toxicity was observed. For the 8 patients treated in remission, 5 remained disease-free after ACT infusion for a median of 42 months (range, 25–82 months). Of the 15 patients treated with relapsed or metastatic disease, 33% had a CR, with 15.4% achieving a PR. Outcome was not statistically influenced by EBV antigen specificity of the infused CTLs or by postinfusion expansion. Both progression-free survival and overall survival were lower in patients with metastatic disease compared with locoregional disease in this study. Investigators concluded that infusion of EBV-CTLs for treatment relapsed/refractory EBV-positive nasopharyngeal carcinoma is safe and provides potential long-term benefit, especially for patients with locoregional disease.44
DIFFERENCES BETWEEN ADULT AND PEDIATRIC SOLID TUMORS: CHALLENGES AND OPPORTUNITIES
Two significant challenges exist for ACT approaches, which do not redirect immune cell specificity in pMST compared with adult solid tumors: (1) differences between adult and pediatric adaptive immune systems and (2) the histologic origin of tumors within the respective age groups. In the adaptive immune system, children have higher levels of regulatory T cells, and their CD4 T cells are biased toward assisting TH2 responses, whereas DC populations are skewed toward a higher plasmacytoid DC–to–myeloid DC ratio.45 The immune system gradually matures through adolescence into a more balanced TH1:TH2 system capable of mounting antipathogen and non–self-tissue responses in adulthood.46 Before this process, the early landscape of infancy is geared toward preventing autoimmune activation at a cost of decreased ability to mount strong T-cell–mediated responses against pathogens and malignant tissue.45,46
Regarding tissue of origin, pediatric cancers generally arise from less-differentiated mesenchymal cell lineage, whereas adult cancers typically derive from more differentiated cells that form components of individual organs.47,48 For example, adult tumors arising from known environmental factors such as UV and cigarette smoking involved in melanoma and lung cancer, respectively, can harbor hundreds of nonsynonymous mutations associated with tumorigenesis.49 Furthermore, DNA repair defects that allow cellular division to occur in cells with potentially mutagenic DNA damage over lengthy periods can have thousands of mutations.50 In contrast, pediatric tumors have an average of 9.6 mutations.47 This lower TMB in pMST also leads to lower immunogenicity and less lymphocytic infiltration.51 Thus, when the source of the ACT is the patient's endogenous T-cell repertoire, there may be fewer tumor-specific T cells present initially. The TME of solid tumors provides additional hurdles to effective ACT in children.52-54 Similar to adult tumors, pediatric tumors maintain an immunosuppressive microenvironment including infiltration with tumor-associated macrophages and myeloid-derived suppressor cells as well as metabolic perturbations that create a TME hostile to proliferation of cytotoxic lymphocytes.55-59
POTENTIAL FOR TILS IN pMST
While research continues to improve upon antibody-based immunotherapy, CAR-T, and TCR therapy, TIL-based ACT is also an attractive candidate therapy for treatment of refractory and advanced-stage solid tumors. Tumor-infiltrating lymphocyte–based ACT involves the ex vivo expansion of tumor-derived T cells from a patient and infusing the expanded cell product back to the patient.7,60 The rationale for this approach is that TILs should recognize the patient's TAA but are suppressed within the TME. By expanding the T cells away from the immunosuppressive TME, they should have greater capacity to mount antitumor immune responses upon infusion back into the patient.60 Unlike toxicities with CAR-T therapy, off-target adverse effects are minimized because TAAs are less likely to be coexpressed on nonmalignant tissue.61 Another important difference between TIL-based ACT compared with gene-modified T-cell therapy is the polyclonal nature of the infusion product, which enables a diverse T-cell response to match the heterogeneity seen in solid tumors, potentially decreasing the risk of antigen loss and immune escape.
Most trials reported have focused on adult metastatic melanoma with a 40% to 50% durable objective response rate and a 10% to 20% CR rate.61-65 Preclinical and early clinical TIL trials targeting adult solid tumors illustrate the potential of this modality.60,65-70 According to ClinicalTrials.gov, there more than 100 actively recruiting TIL-based ACT trials for solid tumors enrolling patients 18 years or older. In contrast, the number of actively recruiting TIL-based ACT trials enrolling patients 18 years or younger is only 3 (NCT03645928, NCT03449108, NCT01955460), and these trials focus on diseases that, with the exception of sarcoma, are rare in pediatrics.
INVESTIGATION OF TILS IN pMST: PRECLINICAL AND EARLY CLINICAL
In neuroblastoma, TILs have been successfully expanded from freshly resected tumors with identified CD8- and CD4-positive T cells (both central memory and effector), as well as a significant number of NK T and γδT cells.71 When isolated TILs were nonreactive against neuroblastoma cell lines, they were then transduced with a CAR-T product targeting GD2, which showed in vitro cytotoxicity. MYCN nonamplified tumors have been found to have significantly more cytotoxic TIL signatures than MYCN amplified.72 Another group established 3 allogeneic neuroblastoma cell lines from different metastatic deposits in the same patient, and TILs seemed to preferentially kill neuroblastoma cells in vitro.73 Tumor-associated lymphocytes obtained from neuroblastoma also seem to have an activated phenotype, especially in the CD8+ T cells.74
Wilms tumor samples have demonstrated a higher concentration of CD8+ T cells in the tumor as compared with the peritumoral area.75 Tumor-infiltrating lymphocytes have also been derived from Wilms tumors that demonstrated cytotoxic activity.76 Another study found that Wilms tumor is infiltrated by immune cells both before and after chemotherapy with elevated NK cells and CD4/CD8+ T cells, which displayed an activated phenotype (HLA-DR, PD1, and CD57 expression increased).77 Germ cell tumors have also been assessed for TILs preclinically: seminomas and germinomas were found to be highly infiltrated by T cells, whereas embryonal carcinomas, choriocarcinomas, and yolk sac tumors demonstrated high infiltration of T cells combined with an immunosuppressive microenvironment.78
For osteosarcoma, T-cell infiltration may correlate with survival; preclinical studies in dogs suggest that there are decreased CD8+ TILs during tumor progression versus regression.79 In a study isolating TILs from 27 patients with pediatric bone tumors (osteosarcoma, ES, giant cell tumor, chondrosarcoma), it was found that only TILs extracted from osteosarcomas were cytotoxic.80 In a study of osteosarcoma of the jaw, 21 samples were analyzed for TILs and positive for CD8+ T cells in approximately half of cases and CD4+ T cells in a third of cases.81 Preclinical data also seem to suggest that exposure to chemotherapy may increase TILs in osteosarcoma. A study of the TARGET database using 27 matched osteosarcoma patient samples assessed changes in the immune environment after chemotherapy and found that neoadjuvant chemotherapy was associated with increased infiltration of all T cells, and specifically CD8+ T cells, into the tumors, with decrease in myeloid-derived suppressive cells.82 Additional work has found that genomic instability of osteosarcoma leads to neoantigen generation, potentially making it more immunogenic or “hot”; in this study, increased TILs were found in metastatic disease compared with the primary tumor, hinting a use for T-cell–based immunotherapy in metastatic osteosarcoma.83 Two active trials using TIL therapy along with lymphodepleting chemotherapy and post-TIL IL-2 are currently enrolling patients as young as 16 years old (NCT03449108, NCT04052334).
Although primarily focused on adults, recent preclinical work has demonstrated the ability to harvest and expand TILs with tumor-specific function in soft tissue sarcoma samples.84 Successful TIL expansion on this study included diagnoses that may affect children and adolescents, including SS, desmoplastic small round cell tumor, and malignant peripheral nerve sheath tumor. Recently, we also demonstrated the ability to harvest and expand TILs from 14 of 18 pMST samples taken from numerous diagnoses including Wilms tumor, neuroblastoma, soft tissue sarcoma, and bone sarcoma, providing preliminary data to support a clinical trial of TIL-based ACT in pMST.85 The work in the adult population supports the currently accruing phase I clinical trial of TIL-based ACT in adolescents and young adults with soft tissue sarcoma (NCT04052334).
OBSTACLES AND FUTURE DIRECTIONS
To optimize the therapeutic potential of TILs in pMST, several obstacles will need to be addressed likely through a combination of ex vivo manipulation of TILs and adjuvant therapeutics.86 First, lack of T-cell recognition of tumor may occur through paucity of tumor-specific neoantigens or through tumor downregulation of major histocompatibility complex molecule or neoantigen expression.87 This may be especially true for pMST that have relatively low TMB. Second, the TME may inhibit optimal TIL activity through multiple mechanisms. Immunosuppressive cells (myeloid-derived suppressor cells, regulatory T cells) may secrete immunosuppressive cytokines (e.g., IL-10, TGF-β) inhibiting TIL function.88,89 The TME may upregulate checkpoint molecules to induce T-cell exhaustion. The coadministration of ICIs alongside TILs is currently being studied in multiple adult solid tumor types to address this. The TME may also impose physical or chemical barriers to the trafficking of TILs to the tumor.90 One strategy to modulate the TME and enhance TIL function includes insertion of a synthetic CD8α:MyD88 coreceptor into TILs during ex vivo expansion conferring improved trafficking to the tumor and resistance to signals inducing T-cell exhaustion.91
CONCLUSIONS
Prior immunotherapy investigation in pediatric cancer has resulted in clinical efficacy in only a small subset of diagnoses, primarily hematologic malignancies. While CAR-T is a transformational therapy for lymphoid malignancy, its success thus far has not translated to pMST. While genetically engineered TCR products can address the paucity of known TAA in pMST and has demonstrated clinical activity in SS, this approach limits the scope of targets within heterogeneous tumors. With the possibility of disease recurrence due to nonimmunogenic tumor cell subclones, TIL-based ACT offers an approach that can limit the refractory response to therapy because of the polyclonal nature of the infusion product. It is therefore an opportune time to fully investigate the utility of TIL-ACT in pMST and also potentially exploit the advantages of targeting multiple TAAs within heterogeneous tumors.
Hurdles presently exist for pMST TIL-based ACT, and differences between adult and pediatric immune systems will require modifications to achieve greater efficacy. Yet, its potential to address unmet needs among refractory, high-tumor-burden and metastatic pMST patients is far too great to overlook. The low presence of mutational burden in the pMST landscape illustrates a need to isolate tumor-reactive T cells from TILs ex vivo away from the suppressive environment within the patient. These isolated T cells can then be reintroduced to initiate an antitumor response likely more reflective of the adult immune system. Another benefit of this strategy is that tumor-reactive T cells expanded in this manner should help circumvent the noted off-target effects of CAR-T ACT. Preclinical work in pMST demonstrates that the hurdle of isolating TILs and expanding them ex vivo to clinically relevant numbers in pMST is certainly surmountable and should undergo further preclinical evaluation as well as clinical investigation in early-phase trials.
Conflicts of Interest and Source of Funding:
J. Mullinax: Moffitt Cancer Center has licensed intellectual property (IP) related to the proliferation and expansion of tumor-infiltrating lymphocytes (TILs) to Iovance Biotherapeutics. He is an inventor on such IP. He participates in sponsored research agreements with Iovance Biotherapeutics, Intellia Therapeutics, and SQZ Biotech, which are not related to this research. He has received research support that is not related to this research from the following entities: NIH-NCI (K08CA252642), Ocala Royal Dames, and V Foundation. He has received ad hoc consulting fees from Merit Medical. S.P.-T.: Moffitt Cancer Center has licensed IP related to the proliferation and expansion of TILs to Iovance Biotherapeutics. Moffitt has also licensed IP to Tuhura Biopharma. She is an inventor on such IP. She is listed as a coinventor on a patent application with Provectus Biopharmaceuticals. She participates in sponsored research agreements with Provectus Biopharmaceuticals, Iovance Biotherapeutics, Intellia Therapeutics, Dyve Biosciences, and Turnstone Biologics, which are not related to this research. She has received research support that is not related to this research from the following entities: NIH-NCI (U01 CA244100-01, R01 CA259387, R43 CA257552-01A1, and R01 CA239219-01A1), Swim Across America, and V Foundation. She has received ad hoc consulting fees from Seagen Inc. and serves as an advisor for KSQ Therapeutics. For the remaining authors, none were declared.
REFERENCES
- 1.Dome JS, Rodriguez-Galindo C, Spunt SL, Santana VM. 92—Pediatric solid tumors. In: Niederhuber JE, Armitage JO, Kastan MB, Doroshow JH, Tepper JE, eds. Abeloff's Clinical Oncology. 6th ed. Philadelphia, PA: Elsevier; 2020:1703–1747.e11. Available at: https://www.sciencedirect.com/science/article/pii/B978032347674400092X. doi: 10.1016/B978-0-323476744.00092-X. Accessed February 22, 2022. [DOI] [Google Scholar]
- 2.Kline NE, Sevier N. Solid tumors in children. J Pediatr Nurs. 2003;18:96–102. doi: 10.1053/jpdn.2003.12. [DOI] [PubMed] [Google Scholar]
- 3.Allen-Rhoades W, Rainusso N. Pediatric solid tumors in children and adolescents: an overview. Pediatr Rev. 2018;39:444–453. doi: 10.1542/pir.2017-0268. [DOI] [PubMed] [Google Scholar]
- 4.Green DM, Kun LE, Matthay KK, et al. Relevance of historical therapeutic approaches to the contemporary treatment of pediatric solid tumors. Pediatr Blood Cancer. 2013;60:1083–1094. doi: 10.1002/pbc.24487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Le TP, Thai TH. The state of cellular adoptive immunotherapy for neuroblastoma and other pediatric solid tumors. Front Immunol. 2017;8:1640. doi: 10.3389/fimmu.2017.01640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Tesfaye M, Savoldo B. Adoptive cell therapy in treating pediatric solid tumors. Curr Oncol Rep. 2018;20:73–79. doi: 10.1007/s11912-018-0715-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Roberts SS, Chou AJ, Cheung NK. Immunotherapy of childhood sarcomas. Front Oncol. 2015;5:181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Brodeur GM. Neuroblastoma: biological insights into a clinical enigma. Nat Rev Cancer. 2003;3:203–216. doi: 10.1038/nrc1014. [DOI] [PubMed] [Google Scholar]
- 9.Maris JM, Hogarty MD, Bagatell R, et al. Neuroblastoma. Lancet. 2007;369:2106–2120. [DOI] [PubMed] [Google Scholar]
- 10.Brivio E, Baruchel A, Beishuizen A, et al. Targeted inhibitors and antibody immunotherapies: novel therapies for paediatric leukaemia and lymphoma. Eur J Cancer. 2022;164:1–17. [DOI] [PubMed] [Google Scholar]
- 11.Yang RK, Sondel PM. Anti-GD2 strategy in the treatment of neuroblastoma. Drugs Future. 2010;35:665. doi: 10.1358/dof.2010.035.08.1513490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Yu AL, Gilman AL, Ozkaynak MF, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med. 2010;363:1324–1334. doi: 10.1056/NEJMoa0911123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Navid F, Sondel PM, Barfield R, et al. Phase I trial of a novel anti-GD2 monoclonal antibody, Hu14.18K322A, designed to decrease toxicity in children with refractory or recurrent neuroblastoma. J Clin Oncol. 2014;32:1445–1452. doi: 10.1200/JCO.2013.50.4423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Voeller J, Sondel PM. Advances in anti-GD2 immunotherapy for treatment of high-risk neuroblastoma. J Pediatr Hematol Oncol. 2019;41:163–169. doi: 10.1097/MPH.0000000000001369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bartholomew J, Washington T, Bergeron S, et al. Dinutuximab: a novel immunotherapy in the treatment of pediatric patients with high-risk neuroblastoma. J Pediatr Oncol Nurs. 2017;34:5–12. doi: 10.1177/1043454216659448. [DOI] [PubMed] [Google Scholar]
- 16.Bailey K, Cost C, Davis I, et al. Emerging novel agents for patients with advanced Ewing sarcoma: a report from the Children's Oncology Group (COG) new agents for Ewing Sarcoma Task Force. F1000Res. 2019;8. doi: 10.12688/f1000research.18139.1 eCollection 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Armideo E, Callahan C, Madonia L. Immunotherapy for high-risk neuroblastoma: management of side effects and complications. J Adv Pract Oncol. 2017;8:44–55. doi: 10.6004/jadpro.2017.8.1.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Tse BC, Navid F, Billups CA, et al. Ocular abnormalities in patients treated with a novel anti-GD2 monoclonal antibody, hu14.18K322A. J AAPOS. 2015;19:112–115. doi: 10.1016/j.jaapos.2014.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hingorani P, Krailo M, Buxton A, et al. Phase II study of antidisialoganglioside antibody, dinutuximab, in combination with GM-CSF in patients with recurrent osteosarcoma (AOST1421): a report from the Children's Oncology Group. J Clin Oncol. 2020;38(suppl 15):10508. [Google Scholar]
- 20.Siebert N, Jensen C, Troschke-Meurer S, et al. Neuroblastoma patients with high-affinity FCGR2A, −3A and stimulatory KIR 2DS2 treated by long-term infusion of anti-GD2 antibody ch14.18/CHO show higher ADCC levels and improved event-free survival. Onco Targets Ther. 2016;5:e1235108. doi: 10.1080/2162402X.2016.1235108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. 2017;377:2500–2501. doi: 10.1056/NEJMc1713444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Choucair K, Morand S, Stanbery L, et al. TMB: a promising immune-response biomarker, and potential spearhead in advancing targeted therapy trials. Cancer Gene Ther. 2020;27:841–853. doi: 10.1038/s41417-020-0174-y. [DOI] [PubMed] [Google Scholar]
- 23.Ma X, Liu Y, Liu Y, et al. Pan-cancer genome and transcriptome analyses of 1,699 paediatric leukaemias and solid tumours. Nature. 2018;555:371–376. doi: 10.1038/nature25795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421. doi: 10.1038/nature12477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Davis KL, Fox E, Merchant MS, et al. Nivolumab in children and young adults with relapsed or refractory solid tumours or lymphoma (ADVL1412): a multicentre, open-label, single-arm, phase 1-2 trial. Lancet Oncol. 2020;21:541–550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Geoerger B, Kang HJ, Yalon-Oren M, et al. Pembrolizumab in paediatric patients with advanced melanoma or a PD-L1-positive, advanced, relapsed, or refractory solid tumour or lymphoma (KEYNOTE-051): interim analysis of an open-label, single-arm, phase 1-2 trial. Lancet Oncol. 2020;21:121–133. [DOI] [PubMed] [Google Scholar]
- 27.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. Lancet Oncol. 2017;18:1493–1501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Naqash AR, O'Sullivan Coyne GH, Moore N, et al. Phase II study of atezolizumab in advanced alveolar soft part sarcoma (ASPS). JCO. 2021;39:11519. doi: 10.1200/JCO.2021.39.15_suppl.11519. [DOI] [Google Scholar]
- 29.Pearson ADJ, Rossig C, Lesa G, et al. ACCELERATE and european medicines agency paediatric strategy forum for medicinal product development of checkpoint inhibitors for use in combination therapy in paediatric patients. Eur J Cancer. 2020;127:52–66. [DOI] [PubMed] [Google Scholar]
- 30.Porter DL, Levine BL, Kalos M, et al. Chimeric antigen receptor–modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365:725–733. doi: 10.1056/NEJMoa1103849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371:1507–1517. doi: 10.1056/NEJMoa1407222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377:2531–2544. doi: 10.1056/NEJMoa1707447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Marayati R, Quinn CH, Beierle EA. Immunotherapy in pediatric solid tumors—a systematic review. Cancers (Basel). 2019;11. doi: 10.3390/cancers11122022 E2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lei W, Xie M, Jiang Q, et al. Treatment-related adverse events of chimeric antigen receptor T-cell (CAR T) in clinical trials: a systematic review and meta-analysis. Cancers (Basel). 2021;13. doi: 10.3390/cancers13153912 3912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Maude SL. Tisagenlecleucel in pediatric patients with acute lymphoblastic leukemia. Clin Adv Hematol Oncol. 2018;16:664–666. [PubMed] [Google Scholar]
- 36.Huang MA, Krishnadas DK, Lucas KG. Cellular and antibody based approaches for pediatric cancer immunotherapy. J Immunol Res. 2015;2015:675269. doi: 10.1155/2015/675269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.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;118:6050–6056. doi: 10.1182/blood-2011-05-354449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.D'Angelo SP, Melchiori L, Merchant MS, et al. Antitumor activity associated with prolonged persistence of adoptively transferred NY-ESO-1 (c259)T cells in synovial sarcoma. Cancer Discov. 2018;8:944–957. doi: 10.1158/2159-8290.CD-17-1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Brennan MF, Antonescu CR, Moraco N, et al. Lessons learned from the study of 10,000 patients with soft tissue sarcoma. Ann Surg. 2014;260:416–421. doi: 10.1097/SLA.0000000000000869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Raza A, Merhi M, Inchakalody VP, et al. Unleashing the immune response to NY-ESO-1 cancer testis antigen as a potential target for cancer immunotherapy. J Transl Med. 2020;18:140–y. doi: 10.1186/s12967-020-02306-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Nowicki TS, Berent-Maoz B, Cheung-Lau G, et al. A pilot trial of the combination of transgenic NY-ESO-1–reactive adoptive cellular therapy with dendritic cell vaccination with or without ipilimumab. Clin Cancer Res. 2019;25:2096–2108. doi: 10.1158/1078-0432.CCR-18-3496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Ramachandran I, Lowther DE, Dryer-Minnerly R, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Mackall CL, Rhee EH, Read EJ, et al. A pilot study of consolidative immunotherapy in patients with high-risk pediatric sarcomas. Clin Cancer Res. 2008;14:4850–4858. doi: 10.1158/1078-0432.CCR-07-4065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Louis CU, Straathof K, Bollard CM, et al. Adoptive transfer of EBV-specific T cells results in sustained clinical responses in patients with locoregional nasopharyngeal carcinoma. J Immunother. 2010;33:983–990. doi: 10.1097/CJI.0b013e3181f3cbf4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Martinez DR, Permar SR, Fouda GG. Contrasting adult and infant immune responses to HIV infection and vaccination. Clin Vaccine Immunol. 2015;23:84–94. doi: 10.1128/CVI.00565-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Simon AK, Hollander GA, McMichael A. Evolution of the immune system in humans from infancy to old age. Proc Biol Sci. 2015;282:20143085. doi: 10.1098/rspb.2014.3085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Rahal Z, Abdulhai F, Kadara H, et al. Genomics of adult and pediatric solid tumors. Am J Cancer Res. 2018;8:1356–1386. [PMC free article] [PubMed] [Google Scholar]
- 48.Vogelstein B, Papadopoulos N, Velculescu VE, et al. Cancer genome landscapes. Science. 2013;339:1546–1558. doi: 10.1126/science.1235122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Govindan R, Ding L, Griffith M, et al. Genomic landscape of non–small cell lung cancer in smokers and never-smokers. Cell. 2012;150:1121–1134. doi: 10.1016/j.cell.2012.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Gryfe R, Gallinger S. Microsatellite instability, mismatch repair deficiency, and colorectal cancer. Surgery. 2001;130:17–20. [DOI] [PubMed] [Google Scholar]
- 51.Casey DL, Cheung NV. Immunotherapy of pediatric solid tumors: treatments at a crossroads, with an emphasis on antibodies. Cancer Immunol Res. 2020;8:161–166. doi: 10.1158/2326-6066.CIR-19-0692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Caruana I, Savoldo B, Hoyos V et al. Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes. Nat Med. 2015;21:524–529. doi: 10.1038/nm.3833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Long AH, Highfill SL, Cui Y, et al. Reduction of MDSCs with all-trans retinoic acid improves CAR therapy efficacy for sarcomas. Cancer Immunol Res. 2016;4:869–880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Long AH, Haso WM, Shern JF, et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med. 2015;21:581–590. doi: 10.1038/nm.3838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kumar V, Patel S, Tcyganov E, et al. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol. 2016;37:208–220. doi: 10.1016/j.it.2016.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Majzner RG, Simon JS, Grosso JF, et al. Assessment of programmed death-ligand 1 expression and tumor-associated immune cells in pediatric cancer tissues. Cancer. 2017;123:3807–3815. doi: 10.1002/cncr.30724. [DOI] [PubMed] [Google Scholar]
- 57.Hutzen B, Ghonime M, Lee J, et al. Immunotherapeutic challenges for pediatric cancers. Mol Ther Oncolytics. 2019;15:38–48. doi: 10.1016/j.omto.2019.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ligon JA, Choi W, Cojocaru G, et al. Pathways of immune exclusion in metastatic osteosarcoma are associated with inferior patient outcomes. J Immunother Cancer. 2021;9:e001772. doi: 10.1136/jitc-001772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Chen L, Oke T, Siegel N, et al. The immunosuppressive niche of soft-tissue sarcomas is sustained by tumor-associated macrophages and characterized by intratumoral tertiary lymphoid structures. Clin Cancer Res. 2020;26:4018–4030. doi: 10.1158/1078-0432.CCR-19-3416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Rosenberg SA, Yang JC, Sherry RM, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 2011;17:4550–4557. doi: 10.1158/1078-0432.CCR-11-0116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hopewell EL, Cox C, Pilon-Thomas S, et al. Tumor-infiltrating lymphocytes: streamlining a complex manufacturing process. Cytotherapy. 2019;21:307–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Besser MJ, Shapira-Frommer R, Itzhaki O, et al. Adoptive transfer of tumor-infiltrating lymphocytes in patients with metastatic melanoma: intent-to-treat analysis and efficacy after failure to prior immunotherapies. Clin Cancer Res. 2013;19:4792–4800. doi: 10.1158/1078-0432.CCR-13-0380. [DOI] [PubMed] [Google Scholar]
- 63.Andersen R, Donia M, Ellebaek E, 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;22:3734–3745. doi: 10.1158/1078-0432.CCR-15-1879. [DOI] [PubMed] [Google Scholar]
- 64.Wu R, Forget MA, Chacon J, et al. Adoptive T-cell therapy using autologous tumor-infiltrating lymphocytes for metastatic melanoma: current status and future outlook. Cancer J. 2012;18:160–175. doi: 10.1097/PPO.0b013e31824d4465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Pilon-Thomas S, Kuhn L, Ellwanger S, et al. Efficacy of adoptive cell transfer of tumor-infiltrating lymphocytes after lymphopenia induction for metastatic melanoma. J Immunother. 2012;35:615–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.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;34:2389–2397. doi: 10.1200/JCO.2016.66.7220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Hall M, Mullinax JE, Royster E, et al. Expansion and characterization of tumor-infiltrating lymphocytes from human sarcoma. J Immunother Cancer. 2015;3(suppl. 2):P19. [Google Scholar]
- 68.Sarnaik A, Hall M, Mullinax J, et al. Clinical results of combined vemurafenib and tumor-infiltrating lymphocyte therapy for metastatic melanoma. J Immunother Cancer. 2015;3(suppl 2):P49. [Google Scholar]
- 69.Liu H, Innamarato PP, Kodumudi K, et al. Intralesional rose bengal in melanoma elicits tumor immunity via activation of dendritic cells by the release of high mobility group box 1. Oncotarget. 2016;7:37893–37905. doi: 10.18632/oncotarget.9247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Mullinax JE, Hall M, Prabhakaran S, et al. Combination of ipilimumab and adoptive cell therapy with tumor-infiltrating lymphocytes for patients with metastatic melanoma. Front Oncol. 2018;8:44. doi: 10.3389/fonc.2018.00044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Olle Hurtado M, Wolbert J, Fisher J, et al. Tumor infiltrating lymphocytes expanded from pediatric neuroblastoma display heterogeneity of phenotype and function. PLoS One. 2019;14:e0216373. doi: 10.1371/journal.pone.0216373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Wei JS, Kuznetsov IB, Zhang S, et al. Clinically relevant cytotoxic immune cell signatures and clonal expansion of T-cell receptors in high-risk MYCN-not-amplified human neuroblastoma. Clin Cancer Res. 2018;24:5673–5684. doi: 10.1158/1078-0432.CCR-18-0599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kataoka Y, Matsumura T, Yamamoto S, et al. Distinct cytotoxicity against neuroblastoma cells of peripheral blood and tumor-infiltrating lymphocytes from patients with neuroblastoma. Cancer Lett. 1993;73:11–21. [DOI] [PubMed] [Google Scholar]
- 74.Carlson LM, De Geer A, Sveinbjornsson B, et al. The microenvironment of human neuroblastoma supports the activation of tumor-associated T lymphocytes. Onco Targets Ther. 2013;2:e23618. doi: 10.4161/onci.23618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Yadav D, Jain Y Dinda A, et al. Tumor-infiltrating lymphocytes in wilms tumor. Indian J Med Paediatr Oncol. 41:34. [Google Scholar]
- 76.Nagai K, Yamada A, Eguchi H, et al. HLA-A2402–restricted and tumor-specific cytotoxic T lymphocytes from tumor-infiltrating lymphocytes of a child with wilms' tumor. Pediatr Res. 1997;42:122–127. doi: 10.1203/00006450-199707000-00019. [DOI] [PubMed] [Google Scholar]
- 77.Holl EK, Routh JC, Johnston AW, et al. Immune expression in children with wilms tumor: a pilot study. J Pediatr Urol. 2019;15:441.e1–441.e8. [DOI] [PubMed] [Google Scholar]
- 78.Boldrini R, De Pasquale MD, Melaiu O, et al. Tumor-infiltrating T cells and PD-L1 expression in childhood malignant extracranial germ-cell tumors. Onco Targets Ther. 2018;8:e1542245. doi: 10.1080/2162402X.2018.1542245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Barber MR, Yang TJ. Tumor infiltrating lymphocytes: CD8+ lymphocytes in canine transmissible venereal sarcomas at different stages of tumor growth. Anticancer Res. 1999;19:1137–1142. [PubMed] [Google Scholar]
- 80.Theoleyre S, Mori K, Cherrier B, et al. Phenotypic and functional analysis of lymphocytes infiltrating osteolytic tumors: use as a possible therapeutic approach of osteosarcoma. BMC Cancer. 2005;5:123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Alves PM, de Arruda JAA, Arantes DAC, et al. Evaluation of tumor-infiltrating lymphocytes in osteosarcomas of the jaws: a multicenter study. Virchows Arch. 2019;474:201–207. doi: 10.1007/s00428-018-2499-6. [DOI] [PubMed] [Google Scholar]
- 82.Deng C, Xu Y, Fu J, et al. Reprograming the tumor immunologic microenvironment using neoadjuvant chemotherapy in osteosarcoma. Cancer Sci. 2020;111:1899–1909. doi: 10.1111/cas.14398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Zhou X, Wu J, Duan C, et al. Retrospective analysis of adoptive TIL therapy plus anti-PD1 therapy in patients with chemotherapy-resistant metastatic osteosarcoma. J Immunol Res. 2020;2020:7890985. doi: 10.1155/2020/7890985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Mullinax JE, Hall M, Beatty M, et al. Expanded tumor-infiltrating lymphocytes from soft tissue sarcoma have tumor-specific function. J Immunother. 2021;44:63–70. doi: 10.1097/CJI.0000000000000355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Metts J, Hensel J, Olmo B, et al. Expansion of tumor-infiltrating lymphocytes and marrow-infiltrating lymphocytes from pediatric malignant solid tumors. In: SITC 2021 annual meeting. 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Qin SS, Melucci AD, Chacon AC, et al. Adoptive T cell therapy for solid tumors: pathway to personalized standard of care. Cell. 2021;10:808. doi: 10.3390/cells10040808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Beatty GL, Gladney WL. Immune escape mechanisms as a guide for cancer immunotherapy. Clin Cancer Res. 2015;21:687–692. doi: 10.1158/1078-0432.CCR-14-1860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Ohue Y, Nishikawa H. Regulatory T (Treg) cells in cancer: can Treg cells be a new therapeutic target? Cancer Sci. 2019;110:2080–2089. doi: 10.1111/cas.14069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 2012;12:253–268. doi: 10.1038/nri3175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.van der Woude LL, Gorris MAJ, Halilovic A, et al. Migrating into the tumor: a roadmap for T cells. Trends Cancer. 2017;3:797–808. [DOI] [PubMed] [Google Scholar]
- 91.Kaczanowska S, Joseph AM, Guo J, et al. A synthetic CD8alpha:MyD88 coreceptor enhances CD8(+) T-cell responses to weakly immunogenic and lowly expressed tumor antigens. Cancer Res. 2017;77:7049–7058. doi: 10.1158/0008-5472.CAN-17-0653. [DOI] [PMC free article] [PubMed] [Google Scholar]