Get 20M+ Full-Text Papers For Less Than $1.50/day. Start a 14-Day Trial for You or Your Team.

Learn More →

Current and Emerging Targets in Immunotherapy for Osteosarcoma

Current and Emerging Targets in Immunotherapy for Osteosarcoma Hindawi Journal of Oncology Volume 2019, Article ID 7035045, 8 pages https://doi.org/10.1155/2019/7035045 Review Article Current and Emerging Targets in Immunotherapy for Osteosarcoma 1 1,2 1 1 Shinji Miwa, Toshiharu Shirai , Norio Yamamoto, Katsuhiro Hayashi, 1 1 1 Akihiko Takeuchi , Kentaro Igarashi, and Hiroyuki Tsuchiya Department of Orthopaedic Surgery, Kanazawa University School of Medicine, Kanazawa 920-8640, Japan Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan Correspondence should be addressed to Toshiharu Shirai; shi-ra-e@med.kanazawa-u.ac.jp Received 29 August 2018; Accepted 4 December 2018; Published 1 January 2019 Academic Editor: Reza Izadpanah Copyright © 2019 Shinji Miwa et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Osteosarcoma is the most common primary malignancy of bone. Although outcomes of patients with osteosarcoma have improved since the introduction of chemotherapy, outcomes of metastatic or unresectable osteosarcomas are still unsatisfactory. To improve osteosarcoma outcomes, the development of novel systemic therapies for osteosarcoma is needed. Since the 1880s, various immunotherapies have been utilized in patients with osteosarcoma and some patients have shown response to the treatment. Based on recent studies about the role of the immune system in malignancies, immunotherapies including immune modulators such as interleukin-2 and muramyl tripeptide, dendritic cells, immune checkpoint inhibitors, and engineered T cells have been utilized in patients with malignancies. Although there are limited reports of immunotherapies for osteosarcoma, immunotherapy is thought to be a promising treatment option for treating osteosarcomas. In this review, an overview of various immunotherapies forosteosarcomaisprovidedandtheirpotential asadjuvant therapiesisdiscussed. 1. Introduction for advanced sarcoma including pazopanib, trabectedin, and eribulin [6], the eca ffi cy of the agents for treating patients Osteosarcoma, the most common primary malignancy of with osteosarcoma remains unclear. er Th efore, novel thera- bone, is thought to originate from mesenchymal stem cells peutic approaches for advanced sarcomas have been sought [1]. Osteosarcoma commonly metastasizes to the lung (more to improve the treatment of osteosarcomas. than 85%) and the bone [2]. Before the introduction of This article reviews immune surveillance for malignancy, chemotherapy, the outcome of patients with osteosarcoma the history of immunotherapy, and recent basic and clin- was poor, and the survival rate of patients with osteosarcoma ical research about immunotherapy for treating osteosar- waslessthan20% before the1970s.Theestablishment coma.Furthermore,wediscuss thefutureperspectivesof of surgical resection with adequate surgical margins and immunotherapy for osteosarcoma treatment. neoadjuvant chemotherapy using methotrexate, doxorubicin, cisplatin, andifosfamideincreased thesurvivalrateupto 2. Role of the Immune System and 60-70% [3]. On the other hand, outcomes of patients with recurrent, metastatic, or unresectable osteosarcomas are still Advancement in Immunotherap unsatisfactory. eTh long-term survival rate for patients with The immune system, a complex organization of immune localizedosteosarcomaisabout 65%,whereasitislessthan cells and mediators, collaborates with other accessory cells 20% for patients with metastatic osteosarcomas [2, 4, 5]. to protect against various pathogens such as viruses and However, no significant improvements have been seen over bacteria.Theinnateimmunesystemconsistsofdendriticcells the last three decades, and recurrent or metastatic osteosar- comas are usually resistant to current standard treatment. (DCs), macrophages, natural killer (NK) cells, neutrophils, Although there are increasing systemic treatment options basophils,andeosinophils.Innateimmunecellsaretheinitial 2 Journal of Oncology defense against foreign antigens (Figure 1). Macrophages and vaccine and inactivated tumor cells can induce antitumor mast cells initiate the inflammatory response by releasing immune responses. cytokines to interact with other immune cells. DCs, strong antigen-presenting cells, play a role in taking foreign antigens 2.2. Macrophage Activator. Muramyl tripeptide (MTP), a and presenting them for recognition by adaptive immune synthetic analog of a component of bacterial cell walls, cells. The adoptive immune cells consist of B lymphocytes, has been developed as a nonspecific immune modula- CD4-positive T helper lymphocytes, and CD8-positive cyto- tor [19]. MTP targets and activates macrophages. Kleiner- toxic T lymphocytes. These cells require direct activation by man reported that treatment with liposome-encapsulated antigen presentation from antigen-presenting cells. Antigen- muramyl tripeptide phosphatidylethanolamine (L-MTP-PE) specific T lymphocytes and B lymphocytes are generated increased levels of TNF-𝛼 and IL-6 [20]. The liposomes by presentation and activation. Furthermore, the innate and encapsulating MTP-PE can deliver the agent selectively to adaptive cells eliminate pathogens and remove damaged cells monocytes and macrophages. After the delivery of MTP-PE [7, 8]. Immunosurveillance for cancer requires the recog- to monocytes and macrophages, these cells become activated nition of tumor-specific antigens, including the products of and tumoricidal [21, 22]. Chemotherapy did not interfere mutated genes, overexpressed normal genes, or genes encod- with L-MTP-PE stimulation of macrophage cytotoxicity in ing viral proteins. In normal conditions, innate immune cells preclinical studies [23]. Meyers reported that adding MTP to and adaptive immune cells detect tumor cells and eliminate conventional chemotherapy improved 6-year overall survival them by activating NK cells, secreting interferons (IFNs), and from 70% to 78% (P = 0.03) in patients with nonmetastatic subsequently activating DCs. However, some tumor cells can osteosarcoma [24]. Furthermore, the risk of death at 6 escape and survive this immune system attack by various years was decreased by 28% for patients receiving MTP. mechanisms including loss of tumor antigens; downregulat- The statistically significant improvement is thought to be ing the major histocompatibility complex (MHC) from the one of the greatest progresses of the treatment of patients surface; altering the tumor microenvironment by recruit- with metastatic osteosarcoma in the last 30 years. On the ing regulatory T cells, myeloid-derived suppressor cells, other hand, a randomized phase 3 trial showed that five- and tumor-associated macrophages; upregulating inhibitory year event-free survival rates for 91 patients with metastatic receptors on T cells, or upregulating inhibitory ligands on osteosarcoma who received L-MTP-PE and those who did tumor cells [9–11]. not were 42% and 26%, respectively [25]. Based on the studies, MTP has been approved by the European Medicine 2.1. Immune Modulator. The stimulation of antitumor Agency for the treatment of patients with osteosarcoma. immune responses by bacterial infection or vaccination has eTh se results suggest a decreased risk of recurrence and been reported for over 100 years (Figure 2). In 1891, Coley metastasis upon treatment with bacterial components in reported that inducing erysipelas and stimulating immune patients with osteosarcoma. reaction by injecting heat-inactivated Streptococcus pyogenes and Serratia marcescens (Coley’s toxin) achieved complete 2.3. Cytokine. Several cytokines have been used for response in approximately 10% of patients with bone and soft immunotherapy in patients with malignancies. IFN-𝛼 tissue sarcomas [12]. In recent studies on the relationship has the ability to induce differentiation and apoptosis, as well between surgical site infection and survival in dogs with as to inhibit proliferation and angiogenesis, and the clinical osteosarcoma, infection had a positive influence on survival efficacy of IFN-𝛼 in several malignancies has been reported [13, 14]. Furthermore, Jeys reported that the 10-year survival [26–29]. In 1977, growth inhibitory effects of IFN- 𝛼 in human for osteosarcoma patients with infection was 85% compared osteosarcoma cells were reported [30]. Furthermore, growth to 63% in patients without infection [15]. These reports inhibitory effects of human IFN- 𝛼 have been reported in of improved survival upon infection suggest that bacteria a mouse model of human osteosarcoma [31]. eTh efficacy have the ability to activate antitumor immune responses. of pegylated IFN-𝛼-2b was investigated in patients with Larsson investigated the effects of immunotherapy using osteosarcoma by an international randomized controlled irradiated tumor cells and the Bacillus Calmette-Guerin trial [32]. The study patients were treated with methotrexate, (BCG) vaccine in a mouse model of osteosarcoma [16]. doxorubicin, and cisplatin (MAP), with or without pegylated In the study, injecting irradiated tumor cells significantly IFN-𝛼-2b. In the study, the 5-year overall survival rates prevented tumor incidence aer ft the injection of living tumor in patients with MAP and MAP + IFN-𝛼-2b were 81% cells, whereas BCG injection had no significant effect on and 84%, respectively. Although the report with a short osteosarcoma. Eilber reported the effect of immunotherapy follow-up period suggests little effect of adjuvant IFN-𝛼-2b, consisting of BCG and an allogenic tumor cell vaccine the follow-up continues to determine long-term survival. [17]. In the study, three of 17 patients (18%) treated with IL-2 induces the activation of lymphocytes and their BCG and tumor vaccine remained alive and disease-free, differentiation into lymphokine-activated killer (LAK) cells whereas 0 of 12 patients without treatment were disease-free. which can recognize and eliminate various tumor cells [33]. Karbach reported that the bacterial vaccine increased levels Although responsestoIL-2havebeenreportedinclinical of immunoregulatory cytokines including interleukin (IL)-6, trials in patients with various malignancies, only clinical tumor necrosis factor (TNF)-𝛼,IFN- 𝛾,and IL1- 𝛽,which trials for renal cell carcinoma and melanoma observed may be involved in inducing tumor regression [18]. Based efficacy of IL-2 [34]. Only a few clinical trials of IL-2 on these studies, it is thought that treatment with a bacterial for sarcomas have been reported [35, 36]. In a clinical Journal of Oncology 3 Figure 1: Interactions between tumor cells and microenvironmen t. Antitumor immune system includes dendritic cells (DCs), CD4 Tcells, CD8 T cells, natural killer (NK) cells, and tumor-suppressing killer B cells. Tumor cells escape immune surveillance by expression of immune checkpoint proteins, regulatory T (Treg) cells, and myeloid-derived suppressor cells (MDSCs). trial using IL-2 with or without reinfusion of LAK for serum IFN-𝛾 levels, reduced pulmonary metastases, and patients with metastatic osteosarcoma, 3-year event-free increased numbers of CD8-positive T lymphocytes in the and overall survival rates were 34% and 45%, respectively metastatic areas [40]. Kawano [41] reported the eeff ct of [35]. Immunotherapy using a monoclonal antibody against combination treatment with tumor lysate-pulsed DCs and an the tumor-associated disialoganglioside GD2, granulocyte anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) antibody macrophage colony-stimulating factor (GM-CSF), and IL- in a mouse model of osteosarcoma. In the study, treatment 2 was associated with a significantly improved outcome as with either the anti-CTLA-4 antibody or tumor lysate- compared with standard treatment in patients with neu- pulsed DCsresultedinanincreased numberof CD8+T roblastoma [36]. In a study of high-dose IL-2 treatment in lymphocytes, inhibition of primary and metastatic lesion relapsed pediatric sarcoma, two of the four patients with growth, prolonged survival, reduced number of regulatory osteosarcoma showed a complete response, although severe T lymphocytes, and increased levels of serum IFN-𝛾,and side effects were observed, including increases in white blood the combination of these treatments enhanced the systemic cells (WBC), creatinine, 𝛾-glutamyltransferase, C-reactive immune response. Furthermore, they reported that combi- protein, glucose, and body weight and decreases of red blood nation treatment with tumor lysate-pulsed DCs and anti- cells, platelets, protein, albumin, and cholinesterase [37]. glucocorticoid-induced tumor necrosis factor receptor (anti- Basedonthe resultsofpaststudies,IFN-𝛼 and IL-2 can acti- GITR) antibodies enhanced the systemic immune response vate antitumor immune responses, although no significant [42]. improvement in osteosarcoma treatment outcomes has been In our phase 1/2 clinical trial of DC-based immunother- observed in the previous trials. apy, 37 patients with bone and soft tissue sarcoma were included in the study [43]. eTh study patients were treated 2.4. Dendritic Cell. Recently, various kinds of cellular with DCs stimulated by tumor lysate, TNF-𝛼,andOK-432. immunotherapy for advanced sarcomas have been developed The patients showed increased levels of IFN- 𝛾 and IL-12 [38]. Treatment of DCs pulsed with tumor lysate significantly without severe toxicity. Among the 35 patients who were increased induction of cytotoxic T lymphocyte (CTL) activity assessed for clinical responses, one patient (3%) showed par- tial response, 6 patients (17%) showed stable disease, and 28 andincreased theserum levelIFN-𝛾 [39]. DCs have been used to enhance tumor-specific immune responses because patients (80%) showed progression of the disease [43]. On the DCs are major antigen-presenting cells initiating cellular other hand, a phase 1 trial using a vaccine of autologous DCs matured with tumor lysate and keyhole limpet hemocyanin immune responses in vivo [40]. DCs pulsed with tumor lysate andreimplantationofcryo-treatedtumorsinducedincreased (KLH) in 13 patients with relapsed osteosarcoma showed 4 Journal of Oncology Immune checkpoint Cytokine therapy: inhibitors IFN-, IL-2 (PD-L, PD-L1, CTLA-4) Immune modulator: Nonspecific immunotherapy Coley toxin, BCG Lymphokine activated killer T cells Dendritic cells Tumor specific immunotherapy Peptide vaccine Genetically modified T cells: (TCR T cells, CAR T cells) 1880 1980 2000 2010 Figure 2: Development of immunotherapy for malignancies. no significant toxicity, although only 2 out of 12 patients therapy for synovial sarcoma using a peptide spanning SYT- exhibited induction of specific T-cell immune response to the SSX fusion region (B peptide) and its HLA-A∗2402 anchor tumor [44]. In a phase 1 study of immunotherapy using tumor substitute, 6 of 12 patients treated with a mixture of the lysate and KLH, 2 of 12 patients with relapsed osteosarcoma peptides had stable disease during the vaccination period, showed induction of specific T cell immune response against although one of the study patients developed intracerebral the tumor although no patients had a clinical response [44]. hemorrhage aer ft the vaccination [50]. In a phase 1 study of immunotherapy using DCs pulsed with tumor lysate, 5 of 10 pediatric patients with solid 2.6. Genetically Modified T Cells. The development of gene- tumors showed progression control and 1 patient showed transfer technology has enabled the genetic transduction of T tumor regression [45]. Merchant et al. reported that 62% of cell receptor (TCR) or chimeric antigen receptor (CAR) into patients with metastatic and/or recurrent pediatric sarcomas conventional T cells. who were treated with immunotherapy using autologous Genes encoding 𝛼 and 𝛽 chains of TCR are introduced lymphocytes, tumor lysate/KLH-pulsed DC vaccinations, into T cells to generate TAA-specicfi TCR T cells [51]. HLA- and/or recombinant human IL7, showed T cell responses A2-restricted TCRs which recognize several antigens, includ- and responders showed prolonged survival period [46]. ing MART-1, gp100, MAGE-A3, and NY-ESO-1, have been In clinical studies of DC-based immunotherapy, no severe cloned [52–55]. TCR cell therapy demonstrated favorable adverse effects have been reported. These studies indicate outcomes in patients with melanoma and synovial sarcoma that the strategy of DC vaccination in relapsed osteosarcoma [56, 57]. Eleven of 18 patients with NY-ESO-1 positive appears safe and resulted in an immunological response in synovialsarcoma(61%) whoreceivedautologousTcells patients with sarcoma, although it resulted in an improved transduced with an NY-ESO-1 reactive TCR demonstrated clinical outcome in only some patients. To improve cellular objective clinical responses, and the vfi e-year survival rate immunotherapy for osteosarcoma, further investigations for was 14% [56]. tumor-associated antigen (TAA) as target of the treatment, Chimeric antigen receptor therapy is an adoptive combination therapy, and favorable indication are needed. immunotherapy utilizing T lymphocytes engineered with chimeric antigen receptors. CAR-T cells can recognize 2.5. Peptide Vaccine. Variousvaccinestargeting tumor tumor antigens in an MHC-independent fashion. Chimeric lysates, proteins, and peptides have been used in clinical trials antigen receptor is composed by an extracellular antigen in patients with sarcoma [47–50]. To eliminate tumor cells, recognition domain, which is called the single-chain variable tumor vaccines have been used to stimulate patients’ immune fragment, and an intracellular signaling domain. CAR-T systems. In treating malignancy, TAA-specific T cells are acti- cell therapy has been used for patients with leukemia vated by antigen-presenting cells as the tumor vaccines are andthisimmunotherapy hasbeenpromising forsarcoma presented on MHC-molecules. Various vaccines have been treatment. A phase 1/2 clinical study using HER2-specific investigated as candidates for vaccine therapy. Tsukahara et CAR-T cells in patients with recurrent/refractory sarcoma al. reported that high expression of papillomavirus binding demonstrated that 4 of 17 patients had stable disease for 12 factor was observed in osteosarcoma cell lines and tumor weeks to 14 months without severe toxicity [58]. On the other tissues [48]. On the other hand, Tsuda et al. reported that hand,CAR-Tcelltherapyisassociatedwithseveraladverse high levels of SART3 were detected in osteosarcoma cell lines effects including cytokine release syndrome and “on-target, and osteosarcoma tissues, and SART3 induced HLA-A24- off-tumor” toxicity, and some of the adverse effects are restricted tumor-specicfi cytotoxic T lymphocytes [49]. In a life-threatening. Cytokine release syndrome is caused by phase 2 trial of four HLA-matched peptides from 31 pooled intensive responses to tumor cell elimination mediated by peptides in 20 patients with refractory bone and soft tissue activated lymphocytes [59]. Excessive levels of cytokines sarcoma, 6 patients had stable disease and 14 patients exhib- including C-reactive protein, IL-6, and IFN-𝛾 are observed in ited disease progression, although no severe adverse effect patients, and high levels cause clinical syndromes including was observed in the study [47]. In a clinical trial of vaccine hypotension, fever, and neurological changes. “On-target, Journal of Oncology 5 off-tumor” toxicity is caused by recognition of tumor- of PD-L1 was correlated with tumor-infiltrating lymphocytes associated antigens on the surface of normal cells, and (TILs)[68].In thestudy,medianoverall survivalforpatients lymphocytes subsequently attack, causing damage to normal with low PD-L1 expression was 89 months compared with tissue. CAR-Tcellsmaykillnormalcellswithtargetantigen 28 months for patients with high PD-L1 expression. Hin- even if the expression of the target antigen is at low level. gorani et al. reported that increased expression of CTLA- eTh refore, CAR-T cell therapy can be applied for cancers 4 in T cells and increased immunosuppressive monocytes with specicfi expression of the antigen. were observed in patients with pediatric sarcoma [69]. A Although further studies of TAAs with high specificity are recent study reported that high levels of PD-1 expression needed for the use of tumor-specific immunotherapies, adop- on peripheral CD4+ and CD8+ T cells were observed in tive cell therapy using TCR or CAR targeting TAAs represents patients with osteosarcoma, and the expression levels of PD-1 a new and promising therapeutic approach for patients with on CD4+ T cells in patients with metastasis were significantly sarcomas. Randomized clinical trials are demanded to assess higher than those without metastasis [66]. Lussier et al. the efficacies and safety of adoptive cell therapies in patients reported that T cells infiltrating PD-L1 antibody-resistant with sarcomas. tumors upregulated inhibitory receptors such as CTLA-4 [65]. Furthermore, combination immunotherapy using anti- 2.7. Immune Checkpoint Inhibitors. Recent studies have CTLA-4 and anti-PD-L1 antibodies improved overall survival in a mouse model of osteosarcoma, whereas no benefit was focused on the association of immune checkpoints with inhibition of the tumor immune system. Although immune observed upon treatment with an anti-CTLA-4 antibody checkpoints are necessary for maintaining self-tolerance alone [65]. Based on a study of immune checkpoints in patients with osteosarcoma, PD-1, PD-L1, and CTLA-4 influ- and for limiting immune responses to prevent autoimmune disorders, immune checkpoints can allow immune tolerance ence osteosarcoma progression. Therefore, immune check- against tumors. CTLA-4 and programmed death receptor- points are promising targets for treating various malignancies 1 (PD-1), the main inhibitory receptors expressed on T including osteosarcoma. The recent studies of osteosarcoma cells, have been considered as an important part in immune indicate that immune checkpoint can be promising treat- ment for patients with osteosarcoma. It is reported that suppression induced by tumor cells, and these molecules are thought to be candidates as new therapeutic targets in var- the expression of checkpoint molecules such as PD-L1 on ious types of advanced malignancies [60]. Activated T cells osteosarcoma cells correlates with metastases and overall normally express PD-1 on the surface and suppress excessive survival [70]. A multicenter, phase 2 trial of the anti-PD-1 immune responses, including autoimmune reactions. PD-L1, mAb pembrolizumab demonstrated that seven of 40 patients the ligand of PD-1, is expressed in various cells including with bone and soft tissue sarcoma (18%) and two of 40 macrophages and tumor cells, and the interaction between patients (5%) had objective responses [71]; treatment-related PD-1 and PD-L1 is usually inu fl enced by tumor tissues. Dhup- serious adverse events occurred in 11% of patients. eTh kar P et al. reported that anit-PD-1 therapy redirecting M2 study included 22 patients with osteosarcoma; one patient macrophages (immunosuppressive and tumor promoting) to (5%) had a partial response, 6 patients (27%) had stable M1 (anti-tumor) resulted in regression of lung metastasis in disease, and 15 patients (68%) exhibited disease progression. mouse model of osteosarcoma [61]. It is considered that immune checkpoints play some roles BlockadeofPD-1byanti-PD-1 nivolumaboranti- in sarcoma development, and immune checkpoints can be PD-L1 BMS-936559 has demonstrated objective responses promising targets for osteosarcoma treatment. Further basic and improved oncological survival in patients with lung and clinical studies will determine the efficacy of immune cancer, melanoma, renal cell cancer, and ovarian cancer checkpoint inhibitors. [62, 63]. Furthermore, ipilimumab, a monoclonal antibody (mAb)targeting CTLA-4,showedsignicfi antimprovement 2.8. Conclusions. Based on recent studies of the tumor of overall survival in patients with metastatic melanomas microenvironment, mechanisms of tumor invasion and [64]. However, no correlation with immune checkpoints has metastasis, antitumor immune system, and immune check- been reported in some types of sarcoma. In sarcomas with points in malignancies, significant improvements in out- high PD-1 expression, PD-1 inhibitors are considered to be come have been seen for some malignancies. Therefore, promising, while tumor antigen-specicfi treatments, such immunotherapy is an increasingly attractive treatment option as dendritic cell-based immunotherapy, can be a candidate in patients with osteosarcoma. Although the introduction treatment in sarcomas with a high expression of tumor- of chemotherapy dramatically improved the outcomes of specific antigen. osteosarcoma treatment, no marked improvement of the Murine studies suggest activity of checkpoint inhibitors treatment for osteosarcoma has been seen in the last three in osteosarcoma [65]. In patients with osteosarcoma, PD-1 decades. eTh main reasons for the lack of development of and PD-L1 levels negatively correlate with prognosis [66]. osteosarcoma treatment include this type of cancer’s rarity, Zheng investigated the efficacy of nivolumab in a mouse heterogeneity, and the lack of discovery of a tumor-specific model of osteosarcoma and reported that nivolumab-treated antigen. For successful osteosarcoma immunotherapy, eluci- mice had significantly fewer metastatic lung lesions, although dation of the condition of immunosurveillance, discovery of nivolumab had no effect on primary tumor volume and tumor-specific antigen for osteosarcoma, and collaborative growth [67]. Shen et al. reported that high expression of PD-L1 was observed in osteosarcoma patients and expression multicenter studies are necessary. 6 Journal of Oncology Conflicts of Interest [14] D. E. Thrall, S. J. Withrow, B. E. Powers et al., “Radiotherapy prior to cortical allograft limb sparing in dogs with osteosar- eTh authors declare that they have no competing interests. coma: A dose response assay,” International Journal of Radiation Oncology ∙ Biology ∙ Physics,vol.18, no.6,pp.1351–1357, 1990. [15] L.M.Jeys,R.J.Grimer,S.R.Carter,R.M.Tillman, and Acknowledgments A. Abudu, “Post operative infection and increased survival in osteosarcoma patients: Are they associated?” Annals of Surgical This work received funding from The Yasuda Medical Foun- Oncology, vol. 14, no. 10, pp. 2887–2895, 2007. dation. [16] S.-E. Larsson, R. Lorentzon, and L. Boquist, “Immunotherapy with irradiated tumour cells and bcg in experimental osteosar- References coma,” Acta Orthopaedica,vol.52,no.5,pp.469–474,1981. [17] F. R. Eilber, C. Townsend, and D. L. Morton, “Osteosarcoma. [1] V. Y. Jo and C. D. M. Fletcher, “WHO classification of soft tissue Results of treatment employing adjuvant immunotherapy,” tumours: an update based on the 2013 (4th) edition,” Pathology, Clinical Orthopaedics and Related Research, vol. 111, pp. 94–100, vol. 46, no. 2, pp. 95–104, 2014. [2] S. S. Bielack, B. Kempf-Bielack, and G. Delling, “Prognostic [18] J. Karbach, A. Neumann, K. Brand et al., “Phase I clinical trial factors in high-grade osteosarcoma of the extremities or trunk: of mixed bacterial vaccine (Coley’s toxins) in patients with NY- an analysis of 1,702 patients treated on neoadjuvant cooperative ESO-1 expressing cancers: immunological effects and clinical osteosarcoma study group protocols,” Journal of Clinical Oncol- activity,” Clinical Cancer Research,vol.18,no.19,pp.5449–5459, ogy,vol.20,no.3,pp.776–790,2002. [3] A. Longhi, C. Errani, M. De Paolis, M. Mercuri, and G. Bacci, [19] T. Asano and E. S. Kleinerman, “Liposome-encapsulated MTP- “Primary bone osteosarcoma in the pediatric age: state of the PE: A novel biologic agent for cancer therapy,” Journal of art,” Cancer Treatment Reviews,vol.32,no.6,pp.423–436,2006. Immunotherapy,vol.14, no.4,pp.286–292,1993. [4] H. Tsuchiya, Y. Kanazawa, M. E. Abdel-Wanis et al., “Eeff ct [20] E. S. Kleinerman, S.-F. Jia, J. Griffin, N. L. Seibel, R. S. Benjamin, of timing of pulmonary metastases identification on prognosis and N. Jaeff , “Phase II study of liposomal muramyl tripeptide in of patients with osteosarcoma: the Japanese Musculoskeletal osteosarcoma: eTh cytokine cascade and monocyte activation Oncology Group study,” Journal of Clinical Oncology,vol.20, following administration,” Journal of Clinical Oncology,vol.10, no. 16, pp. 3470–3477, 2002. no. 8, pp. 1310–1316, 1992. [5] L.Kager,A.Zoubek,U. Pots ¨ chger et al., “Primary metastatic [21] I.J.Fidler,S.Sone,W.E.Fogler,andZ.L.Barnes,“Eradicationof osteosarcoma: presentation and outcome of patients treated on spontaneous metastases and activation of alveolar macrophages neoadjuvant Cooperative Osteosarcoma Study Group proto- by intravenous injection of liposomes containing muramyl cols,” Journal of Clinical Oncology,vol.21, no.10, pp.2011–2018, dipeptide,” Proceedings of the National Acadamy of Sciences of the United States of America,vol.78, no.3,pp. 1680–1684,1981. [6] A. Kawai, K. Yonemori, S. Takahashi, N. Araki, and T. Ueda, [22] E. G. MacEwen, I. D. Kurzman, R. C. Rosenthal et al., “Systemic therapy for soft tissue sarcoma: proposals for the “era Th py for osteosarcoma in dogs with intravenous injection optimal use of pazopanib, trabectedin, and eribulin,” Advances of liposome-encapsulated muramyl tripeptide,” Journal of the in eTh rapy ,vol.34, no.7,pp. 1556–1571,2017. National Cancer Institute,vol.81,no.12, pp.935–938,1989. [7] K.E.deVisser,A.Eichten,andL.M.Coussens, “Paradoxical [23] E.S.Kleinerman,J.S.Snyder,andN.Jaeff ,“Inufl enceof roles of the immune system during cancer development,” Nature chemotherapy administration on monocyte activation by lipo- Reviews Cancer,vol.6,no. 1, pp.24–37,2006. somal muramyl tripeptide phosphatidylethanolamine in chil- [8] C.L.Mackall,M.S.Merchant, andT.J.Fry,“Immune- dren with osteosarcoma,” Journal of Clinical Oncology,vol.9,no. based therapies for childhood cancer,” Nature Reviews Clinical 2, pp. 259–267, 1991. Oncology,vol.11, no.12, pp.693–703,2014. [24] P. A. Meyers, C. L. Schwartz, M. D. Krailo et al., “Osteosarcoma: [9] C. M. Diaz-Montero, J. Finke, and A. J. Montero, “Myeloid- the addition of muramyl tripeptide to chemotherapy improves derived suppressor cells in cancer: therapeutic, predictive, and overall survival–a report from the children’s oncology group,” prognostic implications,” Seminars in Oncology,vol.41, no.2, Journal of Clinical Oncology,vol.26,no.4,pp.633–638,2008. pp.174–184,2014. [25] A. J. Chou, E. S. Kleinerman, M. D. Krailo et al., “Addition [10] D. Laoui, E. Van Overmeire, P. De Baetselier, J. A. Van Ginder- of muramyl tripeptide to chemotherapy for patients with achter, and G. Raes, “Functional relationship between tumor- newly diagnosed metastatic osteosarcoma: a report from the associated macrophages and macrophage colony-stimulating Children’s Oncology Group,” Cancer, vol. 115, no. 22, pp. 5339– factor as contributors to cancer progression,” Frontiers in 5348, 2009. Immunology, vol. 5, no. 5, p. 489, 2014. [26] J.Whelan,D.Patterson,M.Perisoglouetal.,“eTh roleof [11] H. Nishikawa and S. Sakaguchi, “Regulatory T cells in cancer interferons in the treatment of osteosarcoma,” Pediatric Blood immunotherapy,” Current Opinion in Immunology,vol.27,no.1, &Cancer, vol. 54, no. 3, pp. 350–354, 2010. pp.1–7,2014. [27] R.M. Bukowski,C.Tendler,D.Cutler, E.Rose,M.M.Laughlin, [12] W. B. Coley, “Contribution to the knowledge of sarcoma,” and P. Statkevich, “Treating cancer with PEG intron: phar- Annals of Surgery,vol.14,pp.199–220,1891. macokinetic profile and dosing guidelines for an improved interferon-alpha-2b formulation,” Cancer,vol.95, no.2,pp. [13] B.D.X.Lascelles,W.S.Dernell,M.T.Correaetal.,“Improved 389–396, 2002. survival associated with postoperative wound infection in dogs treated with limb-salvage surgery for osteosarcoma,” Annals of [28] J. M. Kirkwood, L. H. Butterfield, A. A. Tarhini, H. Zarour, P. Surgical Oncology,vol.12, no.12, pp.1073–1083,2005. Kalinski, and S. Ferrone, “Immunotherapy of cancer in 2012,” Journal of Oncology 7 CA: A Cancer Journal for Clinicians,vol.62, no.5,pp. 309–335, [44] N. Himoudi, R. Wallace, K. L. Parsley et al., “Lack of T-cell 2012. responses following autologous tumour lysate pulsed dendritic cell vaccination, in patients with relapsed osteosarcoma,” Clini- [29] A. A. Tarhini and J. M. Kirkwood, “How much of a good cal and Translational Oncology,vol.14,no.4,pp. 271–279, 2012. thing? What duration for interferon Alfa-2b adjuvant therapy?” Journal of Clinical Oncology,vol.30,no.31,pp.3773–3776,2012. [45] J. D. Geiger, R. J. Hutchinson, L. F. Hohenkirk et al., “Vaccina- tion of pediatric solid tumor patients with tumor lysate-pulsed [30] H. Strander and S. Einhorn, “Effect of human leukocyte inter- dendritic cells can expand specific T cells and mediate tumor feron on the growth of human osteosarcoma cells in tissue regression,” Cancer Research,vol.61,no.23,pp.8513–8519,2001. culture,” International Journal of Cancer,vol.19, no.4,pp. 468– [46] M. S. Merchant, D. Bernstein, M. Amoako et al., “Adjuvant 473, 1977. immunotherapy to improve outcome in high-risk pediatric [31] O. Brosjo, ¨ H. C. Bauer, L. A. Brostrom ¨ et al., “Influence sarcomas,” Clinical Cancer Research, vol. 22, no. 13, pp. 3182– of human alpha-interferon on four human osteosarcoma 3191, 2016. xenografts in nude mice,” Cancer Research, vol. 45, no. 11, pp. [47] R. Takahashi, Y. Ishibashi, K. Hiraoka et al., “Phase II study of 5598–5602, 1985. personalized peptide vaccination for refractory bone and soft [32] S. S. Bielack, S. Smeland, J. Whelan et al., “Methotrexate, tissue sarcoma patients,” Cancer Science,vol.104,no.10,pp. doxorubicin, and cisplatin (MAP) plus maintenance pegylated 1285–1294, 2013. interferon alfa-2b versus MAP alone in patients with resectable [48] T. Tsukahara, Y. Nabeta, S. Kawaguchi et al., “Identification of high-grade osteosarcoma and good histologic response to pre- human autologous cytotoxic T-lymphocyte-defined osteosar- operative MAP: first results of the EURAMOS-1 good response coma gene that encodes a transcriptional regulator, papillo- randomized controlled trial,” Journal of Clinical Oncology,vol. mavirus binding factor,” Cancer Research,vol.64, no.15, pp. 33, no. 20, pp. 2279–2287, 2015. 5442–5448, 2004. [33] J. B. Stern and K. A. Smith, “Interleukin-2 induction of T-cell G1 [49] N.Tsuda,K.Murayama,H.Ishidaetal.,“Expression ofanewly progression and c-myb expression,” Science,vol.233,no.4760, defined tumor-rejection antigen SART3 in musculoskeletal pp.203–206,1986. tumors and induction of HLA class I-restricted cytotoxic T lym- [34] S. A. Rosenberg, “IL-2: the first effective immunotherapy for phocytes by SART3-derived peptides,” JournalofOrthopaedic human cancer,” The Journal of Immunology ,vol.192,no.12,pp. Research,vol.19,no.3,pp. 346–351,2001. 5451–5458, 2014. [50] S. Kawaguchi, T. Tsukahara, and K. Ida, “SYT-SSX breakpoint [35] C. Meazza, G. Cefalo, M. Massimino et al., “Primary metastatic peptide vaccines in patients with synovial sarcoma: a study from osteosarcoma: results of a prospective study in children given the Japanese Musculoskeletal Oncology Group,” Cancer Science, chemotherapy and interleukin-2,” Medical Oncology,vol.34, no. vol. 103, no. 9, pp. 1625–1630, 2012. 12,p.191,2017. [51] T. N. M. Schumacher, “T-cell-receptor gene therapy,” Nature [36] A. L. Yu, A. L. Gilman, M. F. Ozkaynak et al., “Anti-GD2 Reviews Immunology,vol.2,no. 7,pp.512–519,2002. antibody with GM-CSF, interleukin-2, and isotretinoin for [52] M.S.Hughes,Y.Y.L.Yu,M.E. Dudley et al.,“TransferofaTCR neuroblastoma,” eTh New England Journal of Medicine ,vol.363, gene derived from a patient with a marked antitumor response no. 14, pp. 1324–1334, 2010. conveys highly active T-cell eeff ctor functions,” Human Gene [37] W. Schwinger, V. Klass, M. Benesch et al., “Feasibility of Therapy ,vol.16, no.4,pp. 457–472, 2005. high-dose interleukin-2 in heavily pretreated pediatric cancer [53] R. A. Morgan, M. E. Dudley, Y. Y. L. Yu et al., “High efficiency patients,” Annals of Oncology, vol. 16, no. 7, pp. 1199–1206, 2005. TCR gene transfer into primary human lymphocytes ao ff rds [38] S. Miwa, H. Nishida, and H. Tsuchiya, “Current status of avid recognition of melanoma tumor antigen glycoprotein 100 immunotherapy for sarcomas,” Immunotherapy,vol.9,no.16, and does not alter the recognition of autologous melanoma pp.1331–1338,2017. antigens,” The Journal of Immunology ,vol.171,no.6,pp.3287– [39] Y.-T. He, Q.-M. Zhang, Q.-C. Kou, and B. Tang, “In vitro 3295, 2003. generation of cytotoxic T lymphocyte response using dendritic [54] N. Chinnasamy, J. A. Wargo, Z. Yu et al., “A TCR targeting cell immunotherapy in osteosarcoma,” Oncology Letters,vol.12, the HLA-A*0201-restricted epitope of MAGE-A3 recognizes no. 2, pp. 1101–1106, 2016. multiple epitopes of the MAGE-A antigen superfamily in [40] M. Kawano, H. Nishida, Y. Nakamoto, H. Tsumura, and H. several types of cancer,” eTh Journal of Immunology ,vol.186,no. Tsuchiya, “Cryoimmunologic antitumor effects enhanced by 2,pp.685–696,2011. dendritic cells in osteosarcoma,” Clinical Orthopaedics and [55] Y.Zhao,Z.Zheng,P.F.Robbins, H.T.Khong, S.A.Rosenberg, Related Research,vol.468,no. 5,pp.1373–1383,2010. and R. A. Morgan, “Primary human lymphocytes transduced [41] M. Kawano, I. Itonaga, T. Iwasaki, and H. Tsumura, “Enhance- with NY-ESO-1 antigen-specific TCR genes recognize and kill ment of antitumor immunity by combining anti-cytotoxic diverse human tumor cell lines,” The Journal of Immunology ,vol. T lymphocyte antigen-4 antibodies and cryotreated tumor 174,no.7,pp. 4415–4423,2005. lysate-pulsed dendritic cells in murine osteosarcoma,” Oncology [56] P. F. Robbins, S. H. Kassim, T. L. N. Tran et al., “A pilot trial Reports,vol.29, no.3,pp. 1001–1006,2013. using lymphocytes genetically engineered with an NY-ESO-1- [42] M. Kawano, K. Tanaka, I. Itonaga et al., “Dendritic cells reactive T-cell receptor: Long-term follow-up and correlates combined with anti-GITR antibody produce antitumor effects with response,” Clinical Cancer Research,vol.21,no.5,pp.1019– in osteosarcoma,” Oncology Reports,vol.34,no.4,pp.1995– 1027, 2015. 2001, 2015. [57] P. F.Robbins,R. A. Morgan,S.A.Feldmanetal.,“Tumor [43] S. Miwa, H. Nishida, Y. Tanzawa et al., “Phase 1/2 study of regression in patients with metastatic synovial cell sarcoma and immunotherapy with dendritic cells pulsed with autologous melanoma using genetically engineered lymphocytes reactive tumor lysate in patients with refractory bone and soft tissue with NY-ESO-1,” Journal of Clinical Oncology,vol.29,no.7,pp. sarcoma,” Cancer,vol.123,no. 9,pp.1576–1584,2017. 917–924, 2011. 8 Journal of Oncology [58] N. Ahmed, V. S. Brawley, M. Hegde et al., “Human epider- mal growth factor receptor 2(HER2)-specific chimeric antigen receptor-modified T cells for the immunotherapy of HER2- positive sarcoma,” Journal of Clinical Oncology,vol.33, no.15, pp.1688–1696,2015. [59] M. S. Magee and A. Snook, “Challenges to chimeric antigen receptor (CAR)-T cell therapy for cancer,” Discovery Medicine, vol. 18, no. 100, pp. 265–271, 2014. [60] F. Conforti, L. Pala, V. Bagnardi et al., “Cancer immunotherapy efficacy and patients’ sex: a systematic review and meta- analysis,” The Lancet Oncology ,vol.19,no.6,pp. 737–746, 2018. [61] P. Dhupkar, N. Gordon, J. Stewart, and E. S. Kleinerman, “Anti- PD-1 therapy redirects macrophages from an M2 to an M1 phenotype inducing regression of OS lung metastases,” Cancer Medicine, vol. 7, no. 6, pp. 2654–2664, 2018. [62] S.L. Topalian,F.S.Hodi, J. R. Brahmeretal.,“Safety,activity, and immune correlates of anti-PD-1 antibody in cancer,” The New England Journal of Medicine,vol.366,no. 26,pp. 2443– 2454, 2012. [63] J.R.Brahmer, S.S. Tykodi,L.Q.M.Chowetal.,“Safety and activity of anti-PD-L1 antibody in patients with advanced cancer,” eTh New England Journal of Medicine ,vol.366,no.26, pp. 2455–2465, 2012. [64] F. S. Hodi, S. J. O’Day, and D. F. McDermott, “Improved survival with ipilimumab in patients with metastatic melanoma,” The New England Journal of Medicine,vol.363,no. 8, pp.711–723, [65] D. M. Lussier, J. L. Johnson, P. Hingorani, and J. N. Blattman, “Combination immunotherapy with 𝛼-CTLA-4 and 𝛼-PD- L1 antibody blockade prevents immune escape and leads to complete control of metastatic osteosarcoma,” Journal for ImmunoTherapy of Cancer ,vol.3,no.1,p. 21,2015. [66] W.Zheng,H. Xiao,H.Liu,andY. Zhou,“Expression of programmed death 1 is correlated with progression of osteosar- coma,” APMIS-Acta Pathologica, Microbiologica et Immunolog- ica Scandinavica,vol.123,no.2, pp.102–107,2015. [67] B. Zheng, T. Ren, and Y. Huang, “PD-1 axis expression in musculoskeletal tumors and antitumor effect of nivolumab in osteosarcoma model of humanized mouse,” Journal of Hema- tology Oncology,vol.11, no.1,p.16, 2018. [68] J. K.Shen,G.M.Cote,E. Choy et al.,“Programmedcell death ligand 1 expression in osteosarcoma,” Cancer Immunology Research,vol.2,no.7, pp.690–698, 2014. [69] P. Hingorani, M. L. Maas, M. P. Gustafson et al., “Increased CTLA-4+ T cells and an increased ratio of monocytes with loss of class II (CD14+HLA-DRlo/neg) found in aggressive pediatric sarcoma patients,” Journal for ImmunoTherapy of Cancer ,vol.3, no.1,p.35, 2015. [70] Y. Liao, L. Chen, Y. Feng et al., “Targeting programmed cell death ligand 1 by CRISPR/Cas9 in osteosarcoma cells,” Oncotarget ,vol.8,no. 18,pp. 30276–30287, 2017. [71] H. A. Tawbi, M. Burgess, V. Bolejack et al., “Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial,” The Lancet Oncology ,vol.18, no.11, pp.1493–1501,2017. MEDIATORS of INFLAMMATION The Scientific Gastroenterology Journal of World Journal Research and Practice Diabetes Research Disease Markers Hindawi Hindawi Publishing Corporation Hindawi Hindawi Hindawi Hindawi www.hindawi.com Volume 2018 http://www www.hindawi.com .hindawi.com V Volume 2018 olume 2013 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 International Journal of Journal of Immunology Research Endocrinology Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 Submit your manuscripts at www.hindawi.com BioMed PPAR Research Research International Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 Journal of Obesity Evidence-Based Journal of Journal of Stem Cells Complementary and Ophthalmology International Alternative Medicine Oncology Hindawi Hindawi Hindawi Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2013 Parkinson’s Disease Computational and Behavioural Mathematical Methods AIDS Oxidative Medicine and in Medicine Neurology Research and Treatment Cellular Longevity Hindawi Hindawi Hindawi Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 http://www.deepdyve.com/assets/images/DeepDyve-Logo-lg.png Journal of Oncology Hindawi Publishing Corporation

Current and Emerging Targets in Immunotherapy for Osteosarcoma

Loading next page...
 
/lp/hindawi-publishing-corporation/current-and-emerging-targets-in-immunotherapy-for-osteosarcoma-xxTdls43v6
Publisher
Hindawi Publishing Corporation
Copyright
Copyright © 2019 Shinji Miwa et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
ISSN
1687-8450
eISSN
1687-8469
DOI
10.1155/2019/7035045
Publisher site
See Article on Publisher Site

Abstract

Hindawi Journal of Oncology Volume 2019, Article ID 7035045, 8 pages https://doi.org/10.1155/2019/7035045 Review Article Current and Emerging Targets in Immunotherapy for Osteosarcoma 1 1,2 1 1 Shinji Miwa, Toshiharu Shirai , Norio Yamamoto, Katsuhiro Hayashi, 1 1 1 Akihiko Takeuchi , Kentaro Igarashi, and Hiroyuki Tsuchiya Department of Orthopaedic Surgery, Kanazawa University School of Medicine, Kanazawa 920-8640, Japan Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan Correspondence should be addressed to Toshiharu Shirai; shi-ra-e@med.kanazawa-u.ac.jp Received 29 August 2018; Accepted 4 December 2018; Published 1 January 2019 Academic Editor: Reza Izadpanah Copyright © 2019 Shinji Miwa et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Osteosarcoma is the most common primary malignancy of bone. Although outcomes of patients with osteosarcoma have improved since the introduction of chemotherapy, outcomes of metastatic or unresectable osteosarcomas are still unsatisfactory. To improve osteosarcoma outcomes, the development of novel systemic therapies for osteosarcoma is needed. Since the 1880s, various immunotherapies have been utilized in patients with osteosarcoma and some patients have shown response to the treatment. Based on recent studies about the role of the immune system in malignancies, immunotherapies including immune modulators such as interleukin-2 and muramyl tripeptide, dendritic cells, immune checkpoint inhibitors, and engineered T cells have been utilized in patients with malignancies. Although there are limited reports of immunotherapies for osteosarcoma, immunotherapy is thought to be a promising treatment option for treating osteosarcomas. In this review, an overview of various immunotherapies forosteosarcomaisprovidedandtheirpotential asadjuvant therapiesisdiscussed. 1. Introduction for advanced sarcoma including pazopanib, trabectedin, and eribulin [6], the eca ffi cy of the agents for treating patients Osteosarcoma, the most common primary malignancy of with osteosarcoma remains unclear. er Th efore, novel thera- bone, is thought to originate from mesenchymal stem cells peutic approaches for advanced sarcomas have been sought [1]. Osteosarcoma commonly metastasizes to the lung (more to improve the treatment of osteosarcomas. than 85%) and the bone [2]. Before the introduction of This article reviews immune surveillance for malignancy, chemotherapy, the outcome of patients with osteosarcoma the history of immunotherapy, and recent basic and clin- was poor, and the survival rate of patients with osteosarcoma ical research about immunotherapy for treating osteosar- waslessthan20% before the1970s.Theestablishment coma.Furthermore,wediscuss thefutureperspectivesof of surgical resection with adequate surgical margins and immunotherapy for osteosarcoma treatment. neoadjuvant chemotherapy using methotrexate, doxorubicin, cisplatin, andifosfamideincreased thesurvivalrateupto 2. Role of the Immune System and 60-70% [3]. On the other hand, outcomes of patients with recurrent, metastatic, or unresectable osteosarcomas are still Advancement in Immunotherap unsatisfactory. eTh long-term survival rate for patients with The immune system, a complex organization of immune localizedosteosarcomaisabout 65%,whereasitislessthan cells and mediators, collaborates with other accessory cells 20% for patients with metastatic osteosarcomas [2, 4, 5]. to protect against various pathogens such as viruses and However, no significant improvements have been seen over bacteria.Theinnateimmunesystemconsistsofdendriticcells the last three decades, and recurrent or metastatic osteosar- comas are usually resistant to current standard treatment. (DCs), macrophages, natural killer (NK) cells, neutrophils, Although there are increasing systemic treatment options basophils,andeosinophils.Innateimmunecellsaretheinitial 2 Journal of Oncology defense against foreign antigens (Figure 1). Macrophages and vaccine and inactivated tumor cells can induce antitumor mast cells initiate the inflammatory response by releasing immune responses. cytokines to interact with other immune cells. DCs, strong antigen-presenting cells, play a role in taking foreign antigens 2.2. Macrophage Activator. Muramyl tripeptide (MTP), a and presenting them for recognition by adaptive immune synthetic analog of a component of bacterial cell walls, cells. The adoptive immune cells consist of B lymphocytes, has been developed as a nonspecific immune modula- CD4-positive T helper lymphocytes, and CD8-positive cyto- tor [19]. MTP targets and activates macrophages. Kleiner- toxic T lymphocytes. These cells require direct activation by man reported that treatment with liposome-encapsulated antigen presentation from antigen-presenting cells. Antigen- muramyl tripeptide phosphatidylethanolamine (L-MTP-PE) specific T lymphocytes and B lymphocytes are generated increased levels of TNF-𝛼 and IL-6 [20]. The liposomes by presentation and activation. Furthermore, the innate and encapsulating MTP-PE can deliver the agent selectively to adaptive cells eliminate pathogens and remove damaged cells monocytes and macrophages. After the delivery of MTP-PE [7, 8]. Immunosurveillance for cancer requires the recog- to monocytes and macrophages, these cells become activated nition of tumor-specific antigens, including the products of and tumoricidal [21, 22]. Chemotherapy did not interfere mutated genes, overexpressed normal genes, or genes encod- with L-MTP-PE stimulation of macrophage cytotoxicity in ing viral proteins. In normal conditions, innate immune cells preclinical studies [23]. Meyers reported that adding MTP to and adaptive immune cells detect tumor cells and eliminate conventional chemotherapy improved 6-year overall survival them by activating NK cells, secreting interferons (IFNs), and from 70% to 78% (P = 0.03) in patients with nonmetastatic subsequently activating DCs. However, some tumor cells can osteosarcoma [24]. Furthermore, the risk of death at 6 escape and survive this immune system attack by various years was decreased by 28% for patients receiving MTP. mechanisms including loss of tumor antigens; downregulat- The statistically significant improvement is thought to be ing the major histocompatibility complex (MHC) from the one of the greatest progresses of the treatment of patients surface; altering the tumor microenvironment by recruit- with metastatic osteosarcoma in the last 30 years. On the ing regulatory T cells, myeloid-derived suppressor cells, other hand, a randomized phase 3 trial showed that five- and tumor-associated macrophages; upregulating inhibitory year event-free survival rates for 91 patients with metastatic receptors on T cells, or upregulating inhibitory ligands on osteosarcoma who received L-MTP-PE and those who did tumor cells [9–11]. not were 42% and 26%, respectively [25]. Based on the studies, MTP has been approved by the European Medicine 2.1. Immune Modulator. The stimulation of antitumor Agency for the treatment of patients with osteosarcoma. immune responses by bacterial infection or vaccination has eTh se results suggest a decreased risk of recurrence and been reported for over 100 years (Figure 2). In 1891, Coley metastasis upon treatment with bacterial components in reported that inducing erysipelas and stimulating immune patients with osteosarcoma. reaction by injecting heat-inactivated Streptococcus pyogenes and Serratia marcescens (Coley’s toxin) achieved complete 2.3. Cytokine. Several cytokines have been used for response in approximately 10% of patients with bone and soft immunotherapy in patients with malignancies. IFN-𝛼 tissue sarcomas [12]. In recent studies on the relationship has the ability to induce differentiation and apoptosis, as well between surgical site infection and survival in dogs with as to inhibit proliferation and angiogenesis, and the clinical osteosarcoma, infection had a positive influence on survival efficacy of IFN-𝛼 in several malignancies has been reported [13, 14]. Furthermore, Jeys reported that the 10-year survival [26–29]. In 1977, growth inhibitory effects of IFN- 𝛼 in human for osteosarcoma patients with infection was 85% compared osteosarcoma cells were reported [30]. Furthermore, growth to 63% in patients without infection [15]. These reports inhibitory effects of human IFN- 𝛼 have been reported in of improved survival upon infection suggest that bacteria a mouse model of human osteosarcoma [31]. eTh efficacy have the ability to activate antitumor immune responses. of pegylated IFN-𝛼-2b was investigated in patients with Larsson investigated the effects of immunotherapy using osteosarcoma by an international randomized controlled irradiated tumor cells and the Bacillus Calmette-Guerin trial [32]. The study patients were treated with methotrexate, (BCG) vaccine in a mouse model of osteosarcoma [16]. doxorubicin, and cisplatin (MAP), with or without pegylated In the study, injecting irradiated tumor cells significantly IFN-𝛼-2b. In the study, the 5-year overall survival rates prevented tumor incidence aer ft the injection of living tumor in patients with MAP and MAP + IFN-𝛼-2b were 81% cells, whereas BCG injection had no significant effect on and 84%, respectively. Although the report with a short osteosarcoma. Eilber reported the effect of immunotherapy follow-up period suggests little effect of adjuvant IFN-𝛼-2b, consisting of BCG and an allogenic tumor cell vaccine the follow-up continues to determine long-term survival. [17]. In the study, three of 17 patients (18%) treated with IL-2 induces the activation of lymphocytes and their BCG and tumor vaccine remained alive and disease-free, differentiation into lymphokine-activated killer (LAK) cells whereas 0 of 12 patients without treatment were disease-free. which can recognize and eliminate various tumor cells [33]. Karbach reported that the bacterial vaccine increased levels Although responsestoIL-2havebeenreportedinclinical of immunoregulatory cytokines including interleukin (IL)-6, trials in patients with various malignancies, only clinical tumor necrosis factor (TNF)-𝛼,IFN- 𝛾,and IL1- 𝛽,which trials for renal cell carcinoma and melanoma observed may be involved in inducing tumor regression [18]. Based efficacy of IL-2 [34]. Only a few clinical trials of IL-2 on these studies, it is thought that treatment with a bacterial for sarcomas have been reported [35, 36]. In a clinical Journal of Oncology 3 Figure 1: Interactions between tumor cells and microenvironmen t. Antitumor immune system includes dendritic cells (DCs), CD4 Tcells, CD8 T cells, natural killer (NK) cells, and tumor-suppressing killer B cells. Tumor cells escape immune surveillance by expression of immune checkpoint proteins, regulatory T (Treg) cells, and myeloid-derived suppressor cells (MDSCs). trial using IL-2 with or without reinfusion of LAK for serum IFN-𝛾 levels, reduced pulmonary metastases, and patients with metastatic osteosarcoma, 3-year event-free increased numbers of CD8-positive T lymphocytes in the and overall survival rates were 34% and 45%, respectively metastatic areas [40]. Kawano [41] reported the eeff ct of [35]. Immunotherapy using a monoclonal antibody against combination treatment with tumor lysate-pulsed DCs and an the tumor-associated disialoganglioside GD2, granulocyte anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) antibody macrophage colony-stimulating factor (GM-CSF), and IL- in a mouse model of osteosarcoma. In the study, treatment 2 was associated with a significantly improved outcome as with either the anti-CTLA-4 antibody or tumor lysate- compared with standard treatment in patients with neu- pulsed DCsresultedinanincreased numberof CD8+T roblastoma [36]. In a study of high-dose IL-2 treatment in lymphocytes, inhibition of primary and metastatic lesion relapsed pediatric sarcoma, two of the four patients with growth, prolonged survival, reduced number of regulatory osteosarcoma showed a complete response, although severe T lymphocytes, and increased levels of serum IFN-𝛾,and side effects were observed, including increases in white blood the combination of these treatments enhanced the systemic cells (WBC), creatinine, 𝛾-glutamyltransferase, C-reactive immune response. Furthermore, they reported that combi- protein, glucose, and body weight and decreases of red blood nation treatment with tumor lysate-pulsed DCs and anti- cells, platelets, protein, albumin, and cholinesterase [37]. glucocorticoid-induced tumor necrosis factor receptor (anti- Basedonthe resultsofpaststudies,IFN-𝛼 and IL-2 can acti- GITR) antibodies enhanced the systemic immune response vate antitumor immune responses, although no significant [42]. improvement in osteosarcoma treatment outcomes has been In our phase 1/2 clinical trial of DC-based immunother- observed in the previous trials. apy, 37 patients with bone and soft tissue sarcoma were included in the study [43]. eTh study patients were treated 2.4. Dendritic Cell. Recently, various kinds of cellular with DCs stimulated by tumor lysate, TNF-𝛼,andOK-432. immunotherapy for advanced sarcomas have been developed The patients showed increased levels of IFN- 𝛾 and IL-12 [38]. Treatment of DCs pulsed with tumor lysate significantly without severe toxicity. Among the 35 patients who were increased induction of cytotoxic T lymphocyte (CTL) activity assessed for clinical responses, one patient (3%) showed par- tial response, 6 patients (17%) showed stable disease, and 28 andincreased theserum levelIFN-𝛾 [39]. DCs have been used to enhance tumor-specific immune responses because patients (80%) showed progression of the disease [43]. On the DCs are major antigen-presenting cells initiating cellular other hand, a phase 1 trial using a vaccine of autologous DCs matured with tumor lysate and keyhole limpet hemocyanin immune responses in vivo [40]. DCs pulsed with tumor lysate andreimplantationofcryo-treatedtumorsinducedincreased (KLH) in 13 patients with relapsed osteosarcoma showed 4 Journal of Oncology Immune checkpoint Cytokine therapy: inhibitors IFN-, IL-2 (PD-L, PD-L1, CTLA-4) Immune modulator: Nonspecific immunotherapy Coley toxin, BCG Lymphokine activated killer T cells Dendritic cells Tumor specific immunotherapy Peptide vaccine Genetically modified T cells: (TCR T cells, CAR T cells) 1880 1980 2000 2010 Figure 2: Development of immunotherapy for malignancies. no significant toxicity, although only 2 out of 12 patients therapy for synovial sarcoma using a peptide spanning SYT- exhibited induction of specific T-cell immune response to the SSX fusion region (B peptide) and its HLA-A∗2402 anchor tumor [44]. In a phase 1 study of immunotherapy using tumor substitute, 6 of 12 patients treated with a mixture of the lysate and KLH, 2 of 12 patients with relapsed osteosarcoma peptides had stable disease during the vaccination period, showed induction of specific T cell immune response against although one of the study patients developed intracerebral the tumor although no patients had a clinical response [44]. hemorrhage aer ft the vaccination [50]. In a phase 1 study of immunotherapy using DCs pulsed with tumor lysate, 5 of 10 pediatric patients with solid 2.6. Genetically Modified T Cells. The development of gene- tumors showed progression control and 1 patient showed transfer technology has enabled the genetic transduction of T tumor regression [45]. Merchant et al. reported that 62% of cell receptor (TCR) or chimeric antigen receptor (CAR) into patients with metastatic and/or recurrent pediatric sarcomas conventional T cells. who were treated with immunotherapy using autologous Genes encoding 𝛼 and 𝛽 chains of TCR are introduced lymphocytes, tumor lysate/KLH-pulsed DC vaccinations, into T cells to generate TAA-specicfi TCR T cells [51]. HLA- and/or recombinant human IL7, showed T cell responses A2-restricted TCRs which recognize several antigens, includ- and responders showed prolonged survival period [46]. ing MART-1, gp100, MAGE-A3, and NY-ESO-1, have been In clinical studies of DC-based immunotherapy, no severe cloned [52–55]. TCR cell therapy demonstrated favorable adverse effects have been reported. These studies indicate outcomes in patients with melanoma and synovial sarcoma that the strategy of DC vaccination in relapsed osteosarcoma [56, 57]. Eleven of 18 patients with NY-ESO-1 positive appears safe and resulted in an immunological response in synovialsarcoma(61%) whoreceivedautologousTcells patients with sarcoma, although it resulted in an improved transduced with an NY-ESO-1 reactive TCR demonstrated clinical outcome in only some patients. To improve cellular objective clinical responses, and the vfi e-year survival rate immunotherapy for osteosarcoma, further investigations for was 14% [56]. tumor-associated antigen (TAA) as target of the treatment, Chimeric antigen receptor therapy is an adoptive combination therapy, and favorable indication are needed. immunotherapy utilizing T lymphocytes engineered with chimeric antigen receptors. CAR-T cells can recognize 2.5. Peptide Vaccine. Variousvaccinestargeting tumor tumor antigens in an MHC-independent fashion. Chimeric lysates, proteins, and peptides have been used in clinical trials antigen receptor is composed by an extracellular antigen in patients with sarcoma [47–50]. To eliminate tumor cells, recognition domain, which is called the single-chain variable tumor vaccines have been used to stimulate patients’ immune fragment, and an intracellular signaling domain. CAR-T systems. In treating malignancy, TAA-specific T cells are acti- cell therapy has been used for patients with leukemia vated by antigen-presenting cells as the tumor vaccines are andthisimmunotherapy hasbeenpromising forsarcoma presented on MHC-molecules. Various vaccines have been treatment. A phase 1/2 clinical study using HER2-specific investigated as candidates for vaccine therapy. Tsukahara et CAR-T cells in patients with recurrent/refractory sarcoma al. reported that high expression of papillomavirus binding demonstrated that 4 of 17 patients had stable disease for 12 factor was observed in osteosarcoma cell lines and tumor weeks to 14 months without severe toxicity [58]. On the other tissues [48]. On the other hand, Tsuda et al. reported that hand,CAR-Tcelltherapyisassociatedwithseveraladverse high levels of SART3 were detected in osteosarcoma cell lines effects including cytokine release syndrome and “on-target, and osteosarcoma tissues, and SART3 induced HLA-A24- off-tumor” toxicity, and some of the adverse effects are restricted tumor-specicfi cytotoxic T lymphocytes [49]. In a life-threatening. Cytokine release syndrome is caused by phase 2 trial of four HLA-matched peptides from 31 pooled intensive responses to tumor cell elimination mediated by peptides in 20 patients with refractory bone and soft tissue activated lymphocytes [59]. Excessive levels of cytokines sarcoma, 6 patients had stable disease and 14 patients exhib- including C-reactive protein, IL-6, and IFN-𝛾 are observed in ited disease progression, although no severe adverse effect patients, and high levels cause clinical syndromes including was observed in the study [47]. In a clinical trial of vaccine hypotension, fever, and neurological changes. “On-target, Journal of Oncology 5 off-tumor” toxicity is caused by recognition of tumor- of PD-L1 was correlated with tumor-infiltrating lymphocytes associated antigens on the surface of normal cells, and (TILs)[68].In thestudy,medianoverall survivalforpatients lymphocytes subsequently attack, causing damage to normal with low PD-L1 expression was 89 months compared with tissue. CAR-Tcellsmaykillnormalcellswithtargetantigen 28 months for patients with high PD-L1 expression. Hin- even if the expression of the target antigen is at low level. gorani et al. reported that increased expression of CTLA- eTh refore, CAR-T cell therapy can be applied for cancers 4 in T cells and increased immunosuppressive monocytes with specicfi expression of the antigen. were observed in patients with pediatric sarcoma [69]. A Although further studies of TAAs with high specificity are recent study reported that high levels of PD-1 expression needed for the use of tumor-specific immunotherapies, adop- on peripheral CD4+ and CD8+ T cells were observed in tive cell therapy using TCR or CAR targeting TAAs represents patients with osteosarcoma, and the expression levels of PD-1 a new and promising therapeutic approach for patients with on CD4+ T cells in patients with metastasis were significantly sarcomas. Randomized clinical trials are demanded to assess higher than those without metastasis [66]. Lussier et al. the efficacies and safety of adoptive cell therapies in patients reported that T cells infiltrating PD-L1 antibody-resistant with sarcomas. tumors upregulated inhibitory receptors such as CTLA-4 [65]. Furthermore, combination immunotherapy using anti- 2.7. Immune Checkpoint Inhibitors. Recent studies have CTLA-4 and anti-PD-L1 antibodies improved overall survival in a mouse model of osteosarcoma, whereas no benefit was focused on the association of immune checkpoints with inhibition of the tumor immune system. Although immune observed upon treatment with an anti-CTLA-4 antibody checkpoints are necessary for maintaining self-tolerance alone [65]. Based on a study of immune checkpoints in patients with osteosarcoma, PD-1, PD-L1, and CTLA-4 influ- and for limiting immune responses to prevent autoimmune disorders, immune checkpoints can allow immune tolerance ence osteosarcoma progression. Therefore, immune check- against tumors. CTLA-4 and programmed death receptor- points are promising targets for treating various malignancies 1 (PD-1), the main inhibitory receptors expressed on T including osteosarcoma. The recent studies of osteosarcoma cells, have been considered as an important part in immune indicate that immune checkpoint can be promising treat- ment for patients with osteosarcoma. It is reported that suppression induced by tumor cells, and these molecules are thought to be candidates as new therapeutic targets in var- the expression of checkpoint molecules such as PD-L1 on ious types of advanced malignancies [60]. Activated T cells osteosarcoma cells correlates with metastases and overall normally express PD-1 on the surface and suppress excessive survival [70]. A multicenter, phase 2 trial of the anti-PD-1 immune responses, including autoimmune reactions. PD-L1, mAb pembrolizumab demonstrated that seven of 40 patients the ligand of PD-1, is expressed in various cells including with bone and soft tissue sarcoma (18%) and two of 40 macrophages and tumor cells, and the interaction between patients (5%) had objective responses [71]; treatment-related PD-1 and PD-L1 is usually inu fl enced by tumor tissues. Dhup- serious adverse events occurred in 11% of patients. eTh kar P et al. reported that anit-PD-1 therapy redirecting M2 study included 22 patients with osteosarcoma; one patient macrophages (immunosuppressive and tumor promoting) to (5%) had a partial response, 6 patients (27%) had stable M1 (anti-tumor) resulted in regression of lung metastasis in disease, and 15 patients (68%) exhibited disease progression. mouse model of osteosarcoma [61]. It is considered that immune checkpoints play some roles BlockadeofPD-1byanti-PD-1 nivolumaboranti- in sarcoma development, and immune checkpoints can be PD-L1 BMS-936559 has demonstrated objective responses promising targets for osteosarcoma treatment. Further basic and improved oncological survival in patients with lung and clinical studies will determine the efficacy of immune cancer, melanoma, renal cell cancer, and ovarian cancer checkpoint inhibitors. [62, 63]. Furthermore, ipilimumab, a monoclonal antibody (mAb)targeting CTLA-4,showedsignicfi antimprovement 2.8. Conclusions. Based on recent studies of the tumor of overall survival in patients with metastatic melanomas microenvironment, mechanisms of tumor invasion and [64]. However, no correlation with immune checkpoints has metastasis, antitumor immune system, and immune check- been reported in some types of sarcoma. In sarcomas with points in malignancies, significant improvements in out- high PD-1 expression, PD-1 inhibitors are considered to be come have been seen for some malignancies. Therefore, promising, while tumor antigen-specicfi treatments, such immunotherapy is an increasingly attractive treatment option as dendritic cell-based immunotherapy, can be a candidate in patients with osteosarcoma. Although the introduction treatment in sarcomas with a high expression of tumor- of chemotherapy dramatically improved the outcomes of specific antigen. osteosarcoma treatment, no marked improvement of the Murine studies suggest activity of checkpoint inhibitors treatment for osteosarcoma has been seen in the last three in osteosarcoma [65]. In patients with osteosarcoma, PD-1 decades. eTh main reasons for the lack of development of and PD-L1 levels negatively correlate with prognosis [66]. osteosarcoma treatment include this type of cancer’s rarity, Zheng investigated the efficacy of nivolumab in a mouse heterogeneity, and the lack of discovery of a tumor-specific model of osteosarcoma and reported that nivolumab-treated antigen. For successful osteosarcoma immunotherapy, eluci- mice had significantly fewer metastatic lung lesions, although dation of the condition of immunosurveillance, discovery of nivolumab had no effect on primary tumor volume and tumor-specific antigen for osteosarcoma, and collaborative growth [67]. Shen et al. reported that high expression of PD-L1 was observed in osteosarcoma patients and expression multicenter studies are necessary. 6 Journal of Oncology Conflicts of Interest [14] D. E. Thrall, S. J. Withrow, B. E. Powers et al., “Radiotherapy prior to cortical allograft limb sparing in dogs with osteosar- eTh authors declare that they have no competing interests. coma: A dose response assay,” International Journal of Radiation Oncology ∙ Biology ∙ Physics,vol.18, no.6,pp.1351–1357, 1990. [15] L.M.Jeys,R.J.Grimer,S.R.Carter,R.M.Tillman, and Acknowledgments A. Abudu, “Post operative infection and increased survival in osteosarcoma patients: Are they associated?” Annals of Surgical This work received funding from The Yasuda Medical Foun- Oncology, vol. 14, no. 10, pp. 2887–2895, 2007. dation. [16] S.-E. Larsson, R. Lorentzon, and L. Boquist, “Immunotherapy with irradiated tumour cells and bcg in experimental osteosar- References coma,” Acta Orthopaedica,vol.52,no.5,pp.469–474,1981. [17] F. R. Eilber, C. Townsend, and D. L. Morton, “Osteosarcoma. [1] V. Y. Jo and C. D. M. Fletcher, “WHO classification of soft tissue Results of treatment employing adjuvant immunotherapy,” tumours: an update based on the 2013 (4th) edition,” Pathology, Clinical Orthopaedics and Related Research, vol. 111, pp. 94–100, vol. 46, no. 2, pp. 95–104, 2014. [2] S. S. Bielack, B. Kempf-Bielack, and G. Delling, “Prognostic [18] J. Karbach, A. Neumann, K. Brand et al., “Phase I clinical trial factors in high-grade osteosarcoma of the extremities or trunk: of mixed bacterial vaccine (Coley’s toxins) in patients with NY- an analysis of 1,702 patients treated on neoadjuvant cooperative ESO-1 expressing cancers: immunological effects and clinical osteosarcoma study group protocols,” Journal of Clinical Oncol- activity,” Clinical Cancer Research,vol.18,no.19,pp.5449–5459, ogy,vol.20,no.3,pp.776–790,2002. [3] A. Longhi, C. Errani, M. De Paolis, M. Mercuri, and G. Bacci, [19] T. Asano and E. S. Kleinerman, “Liposome-encapsulated MTP- “Primary bone osteosarcoma in the pediatric age: state of the PE: A novel biologic agent for cancer therapy,” Journal of art,” Cancer Treatment Reviews,vol.32,no.6,pp.423–436,2006. Immunotherapy,vol.14, no.4,pp.286–292,1993. [4] H. Tsuchiya, Y. Kanazawa, M. E. Abdel-Wanis et al., “Eeff ct [20] E. S. Kleinerman, S.-F. Jia, J. Griffin, N. L. Seibel, R. S. Benjamin, of timing of pulmonary metastases identification on prognosis and N. Jaeff , “Phase II study of liposomal muramyl tripeptide in of patients with osteosarcoma: the Japanese Musculoskeletal osteosarcoma: eTh cytokine cascade and monocyte activation Oncology Group study,” Journal of Clinical Oncology,vol.20, following administration,” Journal of Clinical Oncology,vol.10, no. 16, pp. 3470–3477, 2002. no. 8, pp. 1310–1316, 1992. [5] L.Kager,A.Zoubek,U. Pots ¨ chger et al., “Primary metastatic [21] I.J.Fidler,S.Sone,W.E.Fogler,andZ.L.Barnes,“Eradicationof osteosarcoma: presentation and outcome of patients treated on spontaneous metastases and activation of alveolar macrophages neoadjuvant Cooperative Osteosarcoma Study Group proto- by intravenous injection of liposomes containing muramyl cols,” Journal of Clinical Oncology,vol.21, no.10, pp.2011–2018, dipeptide,” Proceedings of the National Acadamy of Sciences of the United States of America,vol.78, no.3,pp. 1680–1684,1981. [6] A. Kawai, K. Yonemori, S. Takahashi, N. Araki, and T. Ueda, [22] E. G. MacEwen, I. D. Kurzman, R. C. Rosenthal et al., “Systemic therapy for soft tissue sarcoma: proposals for the “era Th py for osteosarcoma in dogs with intravenous injection optimal use of pazopanib, trabectedin, and eribulin,” Advances of liposome-encapsulated muramyl tripeptide,” Journal of the in eTh rapy ,vol.34, no.7,pp. 1556–1571,2017. National Cancer Institute,vol.81,no.12, pp.935–938,1989. [7] K.E.deVisser,A.Eichten,andL.M.Coussens, “Paradoxical [23] E.S.Kleinerman,J.S.Snyder,andN.Jaeff ,“Inufl enceof roles of the immune system during cancer development,” Nature chemotherapy administration on monocyte activation by lipo- Reviews Cancer,vol.6,no. 1, pp.24–37,2006. somal muramyl tripeptide phosphatidylethanolamine in chil- [8] C.L.Mackall,M.S.Merchant, andT.J.Fry,“Immune- dren with osteosarcoma,” Journal of Clinical Oncology,vol.9,no. based therapies for childhood cancer,” Nature Reviews Clinical 2, pp. 259–267, 1991. Oncology,vol.11, no.12, pp.693–703,2014. [24] P. A. Meyers, C. L. Schwartz, M. D. Krailo et al., “Osteosarcoma: [9] C. M. Diaz-Montero, J. Finke, and A. J. Montero, “Myeloid- the addition of muramyl tripeptide to chemotherapy improves derived suppressor cells in cancer: therapeutic, predictive, and overall survival–a report from the children’s oncology group,” prognostic implications,” Seminars in Oncology,vol.41, no.2, Journal of Clinical Oncology,vol.26,no.4,pp.633–638,2008. pp.174–184,2014. [25] A. J. Chou, E. S. Kleinerman, M. D. Krailo et al., “Addition [10] D. Laoui, E. Van Overmeire, P. De Baetselier, J. A. Van Ginder- of muramyl tripeptide to chemotherapy for patients with achter, and G. Raes, “Functional relationship between tumor- newly diagnosed metastatic osteosarcoma: a report from the associated macrophages and macrophage colony-stimulating Children’s Oncology Group,” Cancer, vol. 115, no. 22, pp. 5339– factor as contributors to cancer progression,” Frontiers in 5348, 2009. Immunology, vol. 5, no. 5, p. 489, 2014. [26] J.Whelan,D.Patterson,M.Perisoglouetal.,“eTh roleof [11] H. Nishikawa and S. Sakaguchi, “Regulatory T cells in cancer interferons in the treatment of osteosarcoma,” Pediatric Blood immunotherapy,” Current Opinion in Immunology,vol.27,no.1, &Cancer, vol. 54, no. 3, pp. 350–354, 2010. pp.1–7,2014. [27] R.M. Bukowski,C.Tendler,D.Cutler, E.Rose,M.M.Laughlin, [12] W. B. Coley, “Contribution to the knowledge of sarcoma,” and P. Statkevich, “Treating cancer with PEG intron: phar- Annals of Surgery,vol.14,pp.199–220,1891. macokinetic profile and dosing guidelines for an improved interferon-alpha-2b formulation,” Cancer,vol.95, no.2,pp. [13] B.D.X.Lascelles,W.S.Dernell,M.T.Correaetal.,“Improved 389–396, 2002. survival associated with postoperative wound infection in dogs treated with limb-salvage surgery for osteosarcoma,” Annals of [28] J. M. Kirkwood, L. H. Butterfield, A. A. Tarhini, H. Zarour, P. Surgical Oncology,vol.12, no.12, pp.1073–1083,2005. Kalinski, and S. Ferrone, “Immunotherapy of cancer in 2012,” Journal of Oncology 7 CA: A Cancer Journal for Clinicians,vol.62, no.5,pp. 309–335, [44] N. Himoudi, R. Wallace, K. L. Parsley et al., “Lack of T-cell 2012. responses following autologous tumour lysate pulsed dendritic cell vaccination, in patients with relapsed osteosarcoma,” Clini- [29] A. A. Tarhini and J. M. Kirkwood, “How much of a good cal and Translational Oncology,vol.14,no.4,pp. 271–279, 2012. thing? What duration for interferon Alfa-2b adjuvant therapy?” Journal of Clinical Oncology,vol.30,no.31,pp.3773–3776,2012. [45] J. D. Geiger, R. J. Hutchinson, L. F. Hohenkirk et al., “Vaccina- tion of pediatric solid tumor patients with tumor lysate-pulsed [30] H. Strander and S. Einhorn, “Effect of human leukocyte inter- dendritic cells can expand specific T cells and mediate tumor feron on the growth of human osteosarcoma cells in tissue regression,” Cancer Research,vol.61,no.23,pp.8513–8519,2001. culture,” International Journal of Cancer,vol.19, no.4,pp. 468– [46] M. S. Merchant, D. Bernstein, M. Amoako et al., “Adjuvant 473, 1977. immunotherapy to improve outcome in high-risk pediatric [31] O. Brosjo, ¨ H. C. Bauer, L. A. Brostrom ¨ et al., “Influence sarcomas,” Clinical Cancer Research, vol. 22, no. 13, pp. 3182– of human alpha-interferon on four human osteosarcoma 3191, 2016. xenografts in nude mice,” Cancer Research, vol. 45, no. 11, pp. [47] R. Takahashi, Y. Ishibashi, K. Hiraoka et al., “Phase II study of 5598–5602, 1985. personalized peptide vaccination for refractory bone and soft [32] S. S. Bielack, S. Smeland, J. Whelan et al., “Methotrexate, tissue sarcoma patients,” Cancer Science,vol.104,no.10,pp. doxorubicin, and cisplatin (MAP) plus maintenance pegylated 1285–1294, 2013. interferon alfa-2b versus MAP alone in patients with resectable [48] T. Tsukahara, Y. Nabeta, S. Kawaguchi et al., “Identification of high-grade osteosarcoma and good histologic response to pre- human autologous cytotoxic T-lymphocyte-defined osteosar- operative MAP: first results of the EURAMOS-1 good response coma gene that encodes a transcriptional regulator, papillo- randomized controlled trial,” Journal of Clinical Oncology,vol. mavirus binding factor,” Cancer Research,vol.64, no.15, pp. 33, no. 20, pp. 2279–2287, 2015. 5442–5448, 2004. [33] J. B. Stern and K. A. Smith, “Interleukin-2 induction of T-cell G1 [49] N.Tsuda,K.Murayama,H.Ishidaetal.,“Expression ofanewly progression and c-myb expression,” Science,vol.233,no.4760, defined tumor-rejection antigen SART3 in musculoskeletal pp.203–206,1986. tumors and induction of HLA class I-restricted cytotoxic T lym- [34] S. A. Rosenberg, “IL-2: the first effective immunotherapy for phocytes by SART3-derived peptides,” JournalofOrthopaedic human cancer,” The Journal of Immunology ,vol.192,no.12,pp. Research,vol.19,no.3,pp. 346–351,2001. 5451–5458, 2014. [50] S. Kawaguchi, T. Tsukahara, and K. Ida, “SYT-SSX breakpoint [35] C. Meazza, G. Cefalo, M. Massimino et al., “Primary metastatic peptide vaccines in patients with synovial sarcoma: a study from osteosarcoma: results of a prospective study in children given the Japanese Musculoskeletal Oncology Group,” Cancer Science, chemotherapy and interleukin-2,” Medical Oncology,vol.34, no. vol. 103, no. 9, pp. 1625–1630, 2012. 12,p.191,2017. [51] T. N. M. Schumacher, “T-cell-receptor gene therapy,” Nature [36] A. L. Yu, A. L. Gilman, M. F. Ozkaynak et al., “Anti-GD2 Reviews Immunology,vol.2,no. 7,pp.512–519,2002. antibody with GM-CSF, interleukin-2, and isotretinoin for [52] M.S.Hughes,Y.Y.L.Yu,M.E. Dudley et al.,“TransferofaTCR neuroblastoma,” eTh New England Journal of Medicine ,vol.363, gene derived from a patient with a marked antitumor response no. 14, pp. 1324–1334, 2010. conveys highly active T-cell eeff ctor functions,” Human Gene [37] W. Schwinger, V. Klass, M. Benesch et al., “Feasibility of Therapy ,vol.16, no.4,pp. 457–472, 2005. high-dose interleukin-2 in heavily pretreated pediatric cancer [53] R. A. Morgan, M. E. Dudley, Y. Y. L. Yu et al., “High efficiency patients,” Annals of Oncology, vol. 16, no. 7, pp. 1199–1206, 2005. TCR gene transfer into primary human lymphocytes ao ff rds [38] S. Miwa, H. Nishida, and H. Tsuchiya, “Current status of avid recognition of melanoma tumor antigen glycoprotein 100 immunotherapy for sarcomas,” Immunotherapy,vol.9,no.16, and does not alter the recognition of autologous melanoma pp.1331–1338,2017. antigens,” The Journal of Immunology ,vol.171,no.6,pp.3287– [39] Y.-T. He, Q.-M. Zhang, Q.-C. Kou, and B. Tang, “In vitro 3295, 2003. generation of cytotoxic T lymphocyte response using dendritic [54] N. Chinnasamy, J. A. Wargo, Z. Yu et al., “A TCR targeting cell immunotherapy in osteosarcoma,” Oncology Letters,vol.12, the HLA-A*0201-restricted epitope of MAGE-A3 recognizes no. 2, pp. 1101–1106, 2016. multiple epitopes of the MAGE-A antigen superfamily in [40] M. Kawano, H. Nishida, Y. Nakamoto, H. Tsumura, and H. several types of cancer,” eTh Journal of Immunology ,vol.186,no. Tsuchiya, “Cryoimmunologic antitumor effects enhanced by 2,pp.685–696,2011. dendritic cells in osteosarcoma,” Clinical Orthopaedics and [55] Y.Zhao,Z.Zheng,P.F.Robbins, H.T.Khong, S.A.Rosenberg, Related Research,vol.468,no. 5,pp.1373–1383,2010. and R. A. Morgan, “Primary human lymphocytes transduced [41] M. Kawano, I. Itonaga, T. Iwasaki, and H. Tsumura, “Enhance- with NY-ESO-1 antigen-specific TCR genes recognize and kill ment of antitumor immunity by combining anti-cytotoxic diverse human tumor cell lines,” The Journal of Immunology ,vol. T lymphocyte antigen-4 antibodies and cryotreated tumor 174,no.7,pp. 4415–4423,2005. lysate-pulsed dendritic cells in murine osteosarcoma,” Oncology [56] P. F. Robbins, S. H. Kassim, T. L. N. Tran et al., “A pilot trial Reports,vol.29, no.3,pp. 1001–1006,2013. using lymphocytes genetically engineered with an NY-ESO-1- [42] M. Kawano, K. Tanaka, I. Itonaga et al., “Dendritic cells reactive T-cell receptor: Long-term follow-up and correlates combined with anti-GITR antibody produce antitumor effects with response,” Clinical Cancer Research,vol.21,no.5,pp.1019– in osteosarcoma,” Oncology Reports,vol.34,no.4,pp.1995– 1027, 2015. 2001, 2015. [57] P. F.Robbins,R. A. Morgan,S.A.Feldmanetal.,“Tumor [43] S. Miwa, H. Nishida, Y. Tanzawa et al., “Phase 1/2 study of regression in patients with metastatic synovial cell sarcoma and immunotherapy with dendritic cells pulsed with autologous melanoma using genetically engineered lymphocytes reactive tumor lysate in patients with refractory bone and soft tissue with NY-ESO-1,” Journal of Clinical Oncology,vol.29,no.7,pp. sarcoma,” Cancer,vol.123,no. 9,pp.1576–1584,2017. 917–924, 2011. 8 Journal of Oncology [58] N. Ahmed, V. S. Brawley, M. Hegde et al., “Human epider- mal growth factor receptor 2(HER2)-specific chimeric antigen receptor-modified T cells for the immunotherapy of HER2- positive sarcoma,” Journal of Clinical Oncology,vol.33, no.15, pp.1688–1696,2015. [59] M. S. Magee and A. Snook, “Challenges to chimeric antigen receptor (CAR)-T cell therapy for cancer,” Discovery Medicine, vol. 18, no. 100, pp. 265–271, 2014. [60] F. Conforti, L. Pala, V. Bagnardi et al., “Cancer immunotherapy efficacy and patients’ sex: a systematic review and meta- analysis,” The Lancet Oncology ,vol.19,no.6,pp. 737–746, 2018. [61] P. Dhupkar, N. Gordon, J. Stewart, and E. S. Kleinerman, “Anti- PD-1 therapy redirects macrophages from an M2 to an M1 phenotype inducing regression of OS lung metastases,” Cancer Medicine, vol. 7, no. 6, pp. 2654–2664, 2018. [62] S.L. Topalian,F.S.Hodi, J. R. Brahmeretal.,“Safety,activity, and immune correlates of anti-PD-1 antibody in cancer,” The New England Journal of Medicine,vol.366,no. 26,pp. 2443– 2454, 2012. [63] J.R.Brahmer, S.S. Tykodi,L.Q.M.Chowetal.,“Safety and activity of anti-PD-L1 antibody in patients with advanced cancer,” eTh New England Journal of Medicine ,vol.366,no.26, pp. 2455–2465, 2012. [64] F. S. Hodi, S. J. O’Day, and D. F. McDermott, “Improved survival with ipilimumab in patients with metastatic melanoma,” The New England Journal of Medicine,vol.363,no. 8, pp.711–723, [65] D. M. Lussier, J. L. Johnson, P. Hingorani, and J. N. Blattman, “Combination immunotherapy with 𝛼-CTLA-4 and 𝛼-PD- L1 antibody blockade prevents immune escape and leads to complete control of metastatic osteosarcoma,” Journal for ImmunoTherapy of Cancer ,vol.3,no.1,p. 21,2015. [66] W.Zheng,H. Xiao,H.Liu,andY. Zhou,“Expression of programmed death 1 is correlated with progression of osteosar- coma,” APMIS-Acta Pathologica, Microbiologica et Immunolog- ica Scandinavica,vol.123,no.2, pp.102–107,2015. [67] B. Zheng, T. Ren, and Y. Huang, “PD-1 axis expression in musculoskeletal tumors and antitumor effect of nivolumab in osteosarcoma model of humanized mouse,” Journal of Hema- tology Oncology,vol.11, no.1,p.16, 2018. [68] J. K.Shen,G.M.Cote,E. Choy et al.,“Programmedcell death ligand 1 expression in osteosarcoma,” Cancer Immunology Research,vol.2,no.7, pp.690–698, 2014. [69] P. Hingorani, M. L. Maas, M. P. Gustafson et al., “Increased CTLA-4+ T cells and an increased ratio of monocytes with loss of class II (CD14+HLA-DRlo/neg) found in aggressive pediatric sarcoma patients,” Journal for ImmunoTherapy of Cancer ,vol.3, no.1,p.35, 2015. [70] Y. Liao, L. Chen, Y. Feng et al., “Targeting programmed cell death ligand 1 by CRISPR/Cas9 in osteosarcoma cells,” Oncotarget ,vol.8,no. 18,pp. 30276–30287, 2017. [71] H. A. Tawbi, M. Burgess, V. Bolejack et al., “Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial,” The Lancet Oncology ,vol.18, no.11, pp.1493–1501,2017. MEDIATORS of INFLAMMATION The Scientific Gastroenterology Journal of World Journal Research and Practice Diabetes Research Disease Markers Hindawi Hindawi Publishing Corporation Hindawi Hindawi Hindawi Hindawi www.hindawi.com Volume 2018 http://www www.hindawi.com .hindawi.com V Volume 2018 olume 2013 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 International Journal of Journal of Immunology Research Endocrinology Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 Submit your manuscripts at www.hindawi.com BioMed PPAR Research Research International Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 Journal of Obesity Evidence-Based Journal of Journal of Stem Cells Complementary and Ophthalmology International Alternative Medicine Oncology Hindawi Hindawi Hindawi Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2013 Parkinson’s Disease Computational and Behavioural Mathematical Methods AIDS Oxidative Medicine and in Medicine Neurology Research and Treatment Cellular Longevity Hindawi Hindawi Hindawi Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018

Journal

Journal of OncologyHindawi Publishing Corporation

Published: Jan 1, 2019

References