SUMMARY
Soft tissue sarcomas are rare neoplasms, with approximately 9,000 new cases in the United States every year. Unfortunately, there is little progress in the treatment of metastatic soft tissue sarcomas in the past two decades beyond the standard approaches of surgery, chemotherapy, and radiation. Immunotherapy is a modality complementary to conventional therapy,. It is appealing because functional anti-tumor activity could affect both local-regional and systemic disease and act over a prolonged period of time. In this report, we review immunotherapeutic investigative strategies being developed, including several tumor vaccine, antigen vaccine, and dendritic cell vaccine strategies.
Keywords: immunotherapy, dendritic cell, intratumoral, radiation therapy, neoadjuvant, soft tissue sarcoma, cytotoxic T lymphocytes, immune-specific response
Clinical challenges in the treatment of soft-tissue sarcomas
Sarcomas are relatively rare neoplasms, with approximately 9,000 new cases of soft tissue sarcomas in the United States every year. Sarcomas affect all age groups: 15% are found in children younger than 15, and 40% occur after age 55. These tumors arise from smooth and skeletal muscles, tendons, fat, fibrous tissue, synovial tissue, vessels, and nerves. Approximately 60% of soft tissue sarcomas arise in the extremities.
There has been little progress in the treatment of metastatic soft tissue sarcomas over the past two decades. In general, soft tissue sarcomas arising in the extremities or trunk metastasize to the lungs. The standard treatment for these patients is systemic chemotherapy. There is no evidence that combination chemotherapy is more effective than single agents in producing improved overall survival, however, tumor response rates are higher with the combination of doxorubicn and ifosfamide. This drug combination often requires hospitalization for infusion and may produce significant toxicity [1]. In a subset of the patients, metastasectomy or careful observation of asymptomatic lesions can be considered. Resection of lung metastases may be performed if the patient can tolerate the surgery and if the number of metastatic lesions is low. Often, this surgery is performed after chemotherapy, so that responsiveness to chemotherapy can be determined with measurable disease in place. The response rate to combination chemotherapy is 30–60% depending on the agents and doses used, as well as the histology, resulting in a median overall survival of 1 to 2 years. Salvage chemotherapy with gemcitabine and docetaxel at the time of progression can be offered. However, many of the patients on second line therapy eventually succumb to the disease.
Biology and challenges of cancer immunotherapy
During the past decade, intensive studies resulted in the identification of many tumor-associated antigens (TAA) expressed by different neoplasms, including melanoma, cervical, colon, lung, and breast cancers [2]. Several groups, including ours, have demonstrated clearly the induction of an antitumor immune response by immunization of animals with appropriate antigens [3]. The immune cells responsible for tumor cell killing appear to be primarily major histocompatibility complex (MHC)-restricted cytotoxic T lymphocytes (CTL) or CD8+ cells. Such CTLs, via their T cell receptor (TCR) detect target cells by recognizing short peptide fragments of endogenous proteins, which are presented to them in complex with MHC class I molecules on the target cell’s surface. Induction of CTL responses requires effective cooperation between antigen-presenting cells (APCs), T-helper (CD4+) cells, and CTLs. The APCs are responsible for effective presentation of antigen and delivery of co-stimulatory signals to CD4+ and CD8+ cells. The most potent APCs known are dendritic cells (DCs).
Dendritic cells arise from the bone marrow and can be isolated from peripheral blood, lymph nodes, spleen, and skin or can be generated in vitro from progenitors from these sources. Relatively immature DCs have a high potential to acquire, process, and present soluble antigens. They are also capable of taking up apoptotic cells and apoptotic bodies and to then process and effectively present antigens from these apoptotic sources [4,5]. Dendritic cells are the only cells capable of stimulating primary immune responses, including cytotoxic T cell responses [6,7], and so play a crucial role in antitumor immune response. Moreover, they are extremely effective in the stimulation of secondary immune responses. After contact with antigens and activation, DCs migrate to draining lymph nodes where they encounter and activate naïve T cells. Efficient presentation of antigens requires upregulation of MHC class II and co-stimulatory molecules. In vivo, this activation of the APC takes place in tissues after contact with cytokines produced by activated macrophages and T cells, as well as during the direct contact with T cells. It is the DCs, and not tumor cells themselves, now thought to be responsible for the induction of antitumor immunity in a tumor-bearing host [8,9].
The induction of an antitumor immune response that is clinically useful, unfortunately, is seriously limited by the defects in the host immune system, attributable to the cancer cells acting directly or indirectly on the immune cells. Defects in both T cells and DCs have been described in animal tumor models and in cancer patients [10]. We and others recently demonstrated that presentation of antigens by functionally competent DCs may overcome some aspects of this immune deficiency in murine model systems. Frequent (every 4–5 days) administration of DCs that were pulsed with tumor-specific antigens resulted in a substantial antitumor effect [10]. This, and the lack of toxicity observed in DC-based immunotherapy trials suggests that DCs might be useful, even for treatment of patients with advanced cancer. This is very attractive for clinical trial development. For therapeutic cellular immunization, DCs have some ideal features.
However, the mere presence of DCs in the vicinity of tumors is insufficient to induce an antitumor response; even though tumor cells themselves are the best TAA source, and apoptosis can occur during normal development, it is an immunologically weak or even tolerizing environment [14]. The optimal presentation of antigens from tumor cells appears to require two conditions: 1) phagocytosis of apoptotic cells by immature DCs -- to provide peptides for MHC class I and II molecule-related presentation -- and 2) a maturation-inducing signal – this may be delivered by exposure to necrotic tumor cells or their products [15].
Apoptosis, or programmed cell death (distinguished from necrosis), is a complex enzymatic pathway culminating in condensation of chromatin, DNA fragmentation, and death. The apoptotic pathway is an active process that depends on RNA and protein synthesis by the dying cell and can be triggered by a variety of stimuli. Gamma-irradiation induces DNA damage and was one of the first methods found to induce apoptosis in vitro [16]. However, most tumors contain hypoxic cells, and they hypoxic environment can protect against ionizing radiation and subsequently the protected cells can repopulate the tumor after radiation [17]. Thus, γ-radiation may provide a good method of producing apoptotic bodies bearing tumor-derived, for induction of antitumor immune responses using DCs.
Novel immunotherapy combined with standard therapy
During the past couple of years, several groups reported successful results in animal tumor models by combining apoptosis-inducing treatment with intratumoral administration of DCs. Candido et al. demonstrated that injection of DCs into tumors with high rate of spontaneous apoptosis resulted in an antitumor effect [18]. Using a mouse sarcoma tumor model, in our group it was demonstrated that combining γ-radiation with local DC administration into the tumor site resulted in the induction of tumor-specific immune responses, which had a significant antitumor effect [19]. Teitz-Tennenbaum and colleagues reported confirmation of these observations and showed that radiotherapy potentiates the efficacy of intratumoral DC administration [20]. Similar effects were observed when chemotherapy was used as the apoptosis-inducing agen;. Tong et al. showed that combining chemotherapy and intratumoral DC administration leads to a potent antitumor effect [21]. In other work in our group, it was demonstrated that although repeated cycles of chemotherapy have negative cumulative effects on immune system function, chemotherapy does not preclude the generation of potent antitumor immune responses in tumor-bearing mice [22]. Recently, Song and Levy reported that intratumoral injection of DCs after chemotherapy led to complete, long-term regression of mouse lymphoma [23]. These studies have established proof-of-concept and demonstrated the therapeutic potential of this approach of placing dendritic cells and in situ apoptotic tumor to induce significant anticancer response for the host. To test clinically the safety and immunological efficacy of this approach, we initiated pilot clinical trials in sarcoma cancer patients with high risk of recurrence and in prostate cancer patients receiving primary definitive-intent radiotherapy.
Survivin as a surrogate marker of antitumor immune responses
Adequate evaluation of antitumor immune responses is a critical problem in development and assessment of cancer immunotherapeutic strategies [24]. It is clear that adequate evaluation of immune responses to cancer vaccines is impossible without measuring immune response to tumor-specific antigens. Unfortunately, in many tumors, such antigens have not been identified. This is especially true for soft tissue sarcoma. One of the most common approaches to solving this problem is to use whole tumor cell lysates as a source. This approach is feasible in patients with large soft tissue sarcomas, and we have employed it in our clinical trials in sarcoma. However, it would be highly beneficial to complement this analysis using defined antigens specific for the tumor cells. Survivin is a protein that can be very effective in this role.
Survivin is a recently described antiapoptotic protein that belongs to the inhibitor of apoptosis protein (IAP) family. It is a short-lived protein with a half-life of about 30 minutes [25]. Its intracytoplasmic degradation is regulated by a ubiquitin-proteasome pathway, which links peptide products of protein degradation and class I MHC peptides in cell surface MHC-epitope complexes, in different cells. Overexpression of survivin in the cytoplasm of tumor cells and its rapid degradation by proteasome-related mechanism may result in increased expression of survivin epitopes surface in association with MHC class I molecules on the tumor cell surface, thus representing tumor-specific targets for CTL lysis. Immune responses to survivin in cancer patients include the generation of antibodies [26] and the activation of type 1 T helper cells as well as CTL [27,28]. Human HLA-A2-positive DCs pulsed with survivin-derived HLA-A2 matching peptides were shown to induce CD8+ effector T cells against target cells that were transfected with survivin DNA, or against non-tumor targets pulsed with survivin-derived peptide [28]. Furthermore, utilizing a functionally dominant-negative, altered surviving protein as the stimulating protein for immunotherapy, our group showed it is possible to generate a survivin-specific immune response in cells of healthy volunteers and of cancer patients [29].
Expression of survivin at a high level is observed in many common human cancers but not in normal, terminally differentiated adult tissues [25,30–36]. Overexpression of survivin in tumors correlates with more aggressive disease and worse survival [37,38]. This expression of survivin was evaluated specifically in malignant tissue samples from 63 soft tissue sarcoma patients as well as from a panel of tumor cell lines [39,40]. The survivin protein levels were quantified by a novel ELISA and by Western blot analyses. High levels of survivin were detected in tumor samples from more than 75% of patients with stage II and from more than 90% of patients with stage III soft tissue sarcoma [39]. In contrast, none or only weak expression of survivin protein was found in the nonmalignant specimens. As a clinical predictor, elevated survivin content in tumor tissue extracts was a significant and independent negative factor for patients with soft tissue sarcomas [39]. The presence of increased survivin expression and its correlation to prognosis also were confirmed in a variety of soft tissue sarcoma subtypes, including leiomyosarcoma, synovial sarcoma, and pleomorphic liposarcoma [41]. Thus, it can be stated that survivin is overexpressed in a large proportion of cancers, and particularly of soft tissue sarcoma patients. Its defined HLA-A2-bound epitopes are recognized by CTL, and this makes surviving and acquisition of increased titer of anti-survivin CTL) an attractive surrogate marker for evaluation of immune responses in patients with soft tissue sarcoma.
Pre-clinical experiments using mouse tumor models
In preliminary studies using animal tumor models, our group addressed the hypothesis that combining apoptosis-inducing therapy with intratumor administration of DCs can result in a potent antitumor response [19,22]. We used mouse models to prove the concept that DCs injected after γ-irradiation of tumor 1) migrate to the tumor site, 2) engulf apoptotic tumor cells, and 3) induce an antitumor immune response. Two well-characterized mouse sarcoma models carrying defined MHC class I-restricted tumor-specific antigens were used: MethA sarcoma and C3 tumor. MethA is a sarcoma induced by methylcholanthrene, developed in BALB/c mice and passaged as an ascitic tumor. It is a relatively immunogenic tumor that carries a mutant endogenous p53 gene. The C3 sarcoma was obtained by transfecting MEC cells of C57BL/6 mice with EJ-ras and plasmid containing human papillomavirus type 16.
MHC class I restricted peptides (tumor-specific and control) were used to evaluate immune responses in both tumor models. DCs were generated from bone marrow of syngeneic mice using GM-CSF and interleukin-4 (MS-A). MethA or C3 cells (4–5 × 105) were injected subcutaneously into the hind legs of BALB/c or C57BL/6 mice, respectively. Treatment was started on the 13–15th day, when tumors reached at least 5–7 mm in diameter. The γ-irradiation (30 or 50 Gy total) was delivered in 3–5 doses of 10 Gy each, over 2–3 weeks. Apoptosis of tumor cells was verified by TUNEL assay at 3, 18, and 48 hours after γ-irradiation [19]. The γ-irradiation was delivered only to the tumor site, and the rest of body was shielded. DCs were injected intravenously (two-thirds of mice) into the tail vein and subcutaneously (one-third of mice) in close proximity to the tumor 3–4 hours after each dose of irradiation. Mice were sacrificed on the 14–16th day of treatment in order to evaluate immune responses or, when tumors became bulky, for evaluation of an antitumor response.
Using fluorescence labeling, we observed that DCs indeed accumulate at the tumor site after irradiation and they engulf apoptotic tumor cells in situ [19]. In the same study, the immune response to TAA in treated mice was evaluated. Tumors were established subcutaneously, and treatment was started when tumors reached 5–7 mm in diameter. Four independent experiments were performed, with four mouse groups for each: γ irradiation only, DC administration only, both, or neither. Mice were sacrificed 3–4 days after the third irradiation (3 weeks after the start of the treatment), splenocytes were isolated, and the presence of peptide-specific interferon-γ-producing CD8+ T cells was evaluated using ELISPOT assays. Only mice treated with combined therapy demonstrated a significant response to the specific peptide [19]. These results were confirmed in experiments with antigen-specific tetramer staining of CD8+ T cells [19]. Thus, γ-irradiation in combination with administration of DCs induces tumor-specific immune responses in tumor-bearing mice. Further, using ex vivo-generated, transgenic DCs with defective antigen presentation, we demonstrated that these observed effects required cross-presentation by donors’ DCs [19].
To evaluate the antitumor effects of the combined therapy, mice were observed up to 45 days from the start of the treatment and sacrificed when tumors became bulky. Again, in each experiment, the four groups were used: γ irradiation only, DC administration only, both, or neither. Each group included 3–6 mice, and experiments were repeated 3–4 times. MethA sarcoma quickly progressed in non-treated mice. Gamma-radiation and DCs delivered separately only slightly delayed the tumor progression. In contrast, by day 14, ten of twelve mice in the combination treatment rejected the tumors, and growth slowed in the other two. [19].
A more prolonged experiment was performed in the poorly immunogenic C3 tumor model. Mice were treated 5 times with γ-irradiation, or 6 times with DCs, or with both and then observed for 45 days from the start of the treatment. Again, tumor growth was significantly reduced in the combined treatment group; by day 20, 4 out of 10 mice in this group rejected tumors [19]. Thus, these data demonstrate that the combination of local γ-irradiation of tumor and DC administration resulted in the generation TAA-specific CTLs in tumor-bearing mice, and a significant immune response was following the combined treatment.
The effect of combined administration of apoptosis-inducing therapy and DCs was investigated in another experimental model, using chemotherapy as the trigger for immunotherapy [22]. Currently, chemotherapy is the most frequently used treatment for most advanced-stage cancers, with conventional chemotherapy given as several cycles over a relatively long period of time. While itis known that chemotherapy is associated with systemic immune suppression, the salient question is whether this context will still be permissive for induction of an effective anticancer immune response.
To address this question, work in our group was conducted with experiments utilizing mice bearing mammary adenocarcinoma expressing a model tumor antigen, influenza virus HA (DA3-HA), and parental tumor (DA3), without that antigen. The experimental animals received different doses of paclitaxel, with or without additional administration of DCs. Paclitaxel was injected 3 times weekly, and DCs were injected either intravenously or into tumor site 36 hours after each injection of paclitaxel. Apoptosis was measured using Annexin V binding or TUNEL assays. The CD8-mediated response of T cells to HA-derived peptide epitope was measured with ELISPOT and the CD4-mediated response of T cells to HA-derived peptide was measured by a proliferation assay. The combined treatment resulted in induction of HA-specific CD8-mediated response in all 9 tested mice and CD4-mediated responses in 4 of 6 treated mice [22]. These effects were observed only if DCs were injected into tumor site, but not when injected intravenously. No specific responses were found in mice treated with either chemotherapy or DCs alone. Injection of dexamethasone together with paclitaxel (to mimic the frequent usual coadminstration of corticosteroids with taxanes in clinical practice) did not affect the induction of immune responses. A significant antitumor effect with combined treatment was observed both for the DA3-HA or the DA3 tumor [22].
Initial results of pilot clinical trial in soft tissue sarcoma patients
The above murine data and results from other groups have provided the foundatinos for a clinical trial to combine radiation and intratumoral injection of DCs in cancer patients. At Moffitt Cancer Center, we conducted a phase I/II clinical trial in patients with high-grade, large (> 5 cm diameter) extremity soft tissue sarcoma (NCT00365872), a group for whom there about 50% risk of developing recurrent metastatic disease. In addition, these patients generally benefit (in terms of size reduction and potentially smaller tumor) from neoadjuvant external beam radiation therapy (EBRT) to the tumor prior to resection. Goals of the trial were to address he safety of combined administration of EBRT and intratumoral placement of DCs in the neoadjuvant setting, and additionally to evaluated sarcoma-specific immune responses (Figure 1) [42–45].
Figure 1. MCC 14497 Trial Design.

General treatment scheme for cellular dendritic cell (DC) immunotherapy of sarcoma.
Seventeen eligible patients were treated in the neoadjuvant setting with EBRT, combined with four weekly intratumoral injections of 107 DCs followed by complete resection. Patients first underwent leukapheresis to collect peripheral blood mononuclear cells (PBMCs) for the generation of DCs. The PBMCs were stored frozen in aliquots and were thawed 1 week before each injection so that freshly generated DCs would be used. For the EBRT, megavoltage equipment with effective photon energies from 4 to 23 MV was utilized. Any field arrangement that covered the target volume was allowed. All fields were treated every day. The clinical target volume (CTV) was the gross tumor as per planning study(s) plus 5–10 cm longitudinal margin. The planning target volume (PTV) was the CTV plus 2.0 cm. Cerroband blocks or multi-leaf collimators were used to limit the dose to nearby normal structures. The radial CTV could be limited in order to spare a required strip of skin (2–3 cm) but had to cover the tumor with a minimum of 2 cm. The PTV was treated to 50.4 Gy in 1.8-Gy fractions (total of 28 fractions), given five times per week. The prescribed dose was defined on or near the central axis. The PTV was covered by the 95% isodose line; a significant hot spot did not exceed 110%.
One week after the start of radiation therapy, the autologous PBMCs were thawed, and DCs were generated using serum-free X-VIVO-15 medium, interleukin-4, and GM-CSF. In preparation for this trial, we optimized the conditions for DC generation using serum-free medium and clinical-grade growth factor and cytokines. The radiation treatment was started on Monday; DCs were injected on Fridays 2–4 hours after that day’s radiation fraction. The 3-day interval between the DC injection and the following Monday’s dose of radiation allowed DCs to engulf apoptotic tumor cells and migrate out of the tumor site into draining lymph nodes. Injections of DCs were performed after the 10th, 15th, and 20th doses of radiation. Biopsies of the tumor area were obtained before EBRT, after the second DC injection, and at completion of surgical resection. Blood was collected for immunological testing initially, after the second DC injection, and in follow-up at 2-month intervals to monitor the longevity of the response [42–45].
To date, all patients have completed treatment and full immunologic analysis. The treatment was well tolerated with no unexpected, serious adverse events occurring. No patient had assay-detected tumor-specific immune responses before combined EBRT/DC therapy; 9 patients (52.9%) developed tumor-specific immune responses, which lasted from 11 to 42 weeks. Interestingly, this response became progressively stronger throughout the course of treatment (in some cases a 4-fold increase over control) after the last DC injection. Furthermore, these responses persisted even 30 weeks after the treatment start. Twelve of 17 patients were progression free after 1 year, (71%, 95% CI 47–87%). Histologic evaluation demonstrated that treatment caused a dramatic accumulation of T cells in the tumor. The presence of CD4+ T cells in the tumor positively correlated with tumor-specific immune responses that developed following combined therapy. Accumulation of myeloid-derived suppressor cells negatively correlated with the development of tumor-specific immune responses, but for regulatory T cells, it was not the case. Experiments with 111I-labeled DCs demonstrated that these APCs need at least 48 hours to start migrating from tumor site. Thus, we concluded that the combination of intratumoral DC administration and EBRT is safe and results in induction of antitumor immune responses. Indeed, this therapy is promising and needs further testing in clinical trials designed to assess clinical efficacy [42–45].
Initial results of pilot clinical trial in synovial cell sarcoma patients
A separate clinical trial was performed using genetically engineered lymphocytes reactive with antigen for sarcoma in patients with metastatic synovial cell sarcoma refractory to all standard treatments [2]. Patients with tumors positive for the NY-ESO-1 antigen were treated with autologous T cells that had been transduced with the corresponding TCR specific for peptide of that antigen, and also treated with interleukin-2 at a dose of 720,000 IU/kg of interleukin-2, up to tolerance, with the immunotherapy given following after preparative chemotherapy. Objective clinical responses were evaluated using Response Evaluation Criteria in Solid Tumors (RECIST). Objective clinical responses were observed in 4 of 6 patients with synovial cell sarcoma and in 5 of 11 patients with melanoma bearing tumors expressing NY-ESO-1. Two of 11 patients with melanoma demonstrated complete regression that persisted after 1 year. A partial response lasting 18 months was observed in one patient with synovial cell sarcoma. These observations indicate that TCR-based gene therapy directed against NY-ESO-1 is a viable therapeutic approach to develop for patients with synovial cell sarcoma [2].
Future perspective
Cellular immunotherapy is a novel in treatment for sarcoma. The intratumoral placement of ex vivo-generated, autologous DCs in concert with apoptosis induction can generate systemic immunity in murine systems and is feasible and promising in an early clinical trial. In addition, TCR-based gene therapy directed against a known sarcoma antigen bears interest for treating a nonmelanoma tumor using TCR-transduced T cells. In the clinical setting a successful induction of systemic antitumor immunity may provide substantial clinical benefit. Our approach, with autologous dendritic cells and in situ tumor apoptosis as the antigen source presents an advantage of eliminating the cumbersome requirement for patient selection based on MHC class I type in future clinical development. Similarly, this approach eliminates patient selection based on expression of defined TAAs and reduces the problem of TAA loss during tumor progression. Thus, we believe our approach could dramatically accelerate developing immune-based treatments, as compared to those immunotherapies directed at specific TAAs.
Our group has already demonstrated that the combination of intratumoral administration of DCs with local radiation can generate antitumor immune responses in a pilot study involving soft tissue sarcoma patients possessing localized disease with high-risk features. We have embarked in confirming these observations in a larger, multi-institutional phase II trial with a 10-Gy boost of radiation, which is currently enrolling patients (NCT01347034). Use of TCR-based gene therapies directed against sarcoma antigens continues as well. Future work, and potential integration with acellular immunotherapeutics such as immune checkpoint inhibitors [46] and modulators of dendritic cell phenotype [47] will more importantly define the best ways that this therapy can be translated into impact most meaningfully on the prognosis of soft tissue sarcoma.
EXECUTIVE SUMMARY.
Clinical challenges in the treatment of soft-tissue sarcomas
Little progress has been made in the treatment of metastatic soft tissue sarcomas; development of new options is urgent.
Biology and challenges of cancer immunotherapy
Tumor-associated antigens and induction of an antitumor immune response by immunization with these antigens have been demonstrated
Dendritic cells (DCs) are the most potent antigen presenting cells, capable of taking-up apoptotic cells and processing and presenting their antigens.
Gamma-radiation provides a good method of producing tumor-specific antigens, as apoptotic bodies, for induction of antitumor immune responses using DCs.
Novel immunotherapy combined with standard therapy
In animal tumor models, we have shown that combining apoptosis-inducing therapy with intratumoral administration of DCs results in a potent antitumor immune response.
DCs play a crucial role in antitumor immune responses. Their use in combination with apoptosis-inducing radiotherapy has been demonstrated pre-clinically. Thus, translational clinical testing of this immunotherapeutic strategy is warranted.
Initial results of pilot clinical trial in soft tissue sarcoma patients
We have conducted a phase I/II clinical trial in large, high-grade, soft tissue sarcomas, known to have a 50% risk of developing metastatic disease and to benefit from neoadjuvant external beam radiation therapy [EBRT]. The goal of this trial was to evaluate the safety of combined administration of EBRT and intratumoral placement of DCs in the neoadjuvant setting
Seventeen patients were enrolled. The treatment was well tolerated. Nine patients (52.9%) developed tumor-specific immune responses which positively correlated with the dramatic accumulation of T cells observed in the tumor. Accumulation of myeloid-derived suppressor cells negatively correlated with the development of tumor-specific immune responses, but accumulation of regulatory T cells did not.
The combination of intratumoral DC administration and EBRT is safe, results in the induction of strong antitumor immune response that are promising and warrants further testing in clinical trials designed to assess clinical efficacy
Future perspective
Other strategies to elicit an enhanced antitumor immune responses such as utilizing a local radiotherapy boost or genetically engineered lymphocytes reactive to specific TAAs, are also being evaluated, with preliminary clinical success. Inhibitors of immunologic checkpoints are of contemporary interest in cancer as well.
Footnotes
Conflict of Interest Statement: none of the authors have a conflict of interest.
FINANCIAL DISCLOSURE
No financial relationships to disclose
Contributor Information
Steven Eric Finkelstein, Email: sfinkels@rtsx.com, 21st Century Oncology, Director, Translational Research Consortium, Scottsdale, AZ, Phone: 480-247-0610
Mayer Fishman, Email: Mayer.fishman@moffitt.org, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, Phone: 813-745-8311.
Anthony P. Conley, Email: Anthony.conley@moffitt.org, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, Phone: 813-745-6161.
Dmitry Gabrilovich, Email: Dmitry.gabrilovich@moffitt.org, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, Phone: 813-745-6863.
Scott Antonia, Email: Scott.antonia@moffitt.org, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, Phone: 813-745-3883.
Alberto Chiappori, Email: alberto.chiappori@moffitt.org, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 12902 Magnolia Drive, Tampa, FL 33612, Phone: 813-745-3050
References
Papers of special note have been highlighted as:
* of interest
** of considerable interest
- 1.Patel SR, Vadhan-Raj S, Burgess MA, et al. Results of two consecutive trials of dose-intensive chemotherapy with doxorubicin and ifosfamide in patients with sarcomas. Am J Clin Oncol. 1998;21(3):317–321. doi: 10.1097/00000421-199806000-00025. [DOI] [PubMed] [Google Scholar]
- 2**.Robbins PF, Morgan RA, Feldman SA, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol. 2011;29(7):917–924. doi: 10.1200/JCO.2010.32.2537. TCR-based gene therapies directed against NY-ESO-1 may represent a new and effective therapeutic approach for patients with melanoma and synovial cell sarcoma. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Gabrilovich DI. Dendritic cell vaccines for cancer treatment. Curr Opin Mol Ther. 2002;4(5):452–458. [PubMed] [Google Scholar]
- 4.Albert ML, Pearce SF, Francisco LM, et al. Immature dendritic cells phagocytose apoptotic cells via alphavbeta5 and CD36, and cross-present antigens to cytotoxic T lymphocytes. J Exp Med. 1998;188(7):1359–1368. doi: 10.1084/jem.188.7.1359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5*.Albert ML, Sauter B, Bhardwaj N. Dendritic cells acquire antigen from apoptotic cells and induce class I-restricted CTLs. Nature. 1998;392(6671):86–89. doi: 10.1038/32183. Human dendritic cells, efficiently present antigens derived from apoptotic cells, stimulating class I-restricted CD8+ CTLs and suggesting a mechanism by which potent APCs acquire tumor antigens for stimulation or tolerization of CTLs. [DOI] [PubMed] [Google Scholar]
- 6.Banchereau J, Briere F, Caux C, et al. Immunobiology of dendritic cells. Annu Rev Immunol. 2000;18:767–811. doi: 10.1146/annurev.immunol.18.1.767. [DOI] [PubMed] [Google Scholar]
- 7.Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392(6673):245–252. doi: 10.1038/32588. [DOI] [PubMed] [Google Scholar]
- 8.Huang AY, Golumbek P, Ahmadzadeh M, Jaffee E, Pardoll D, Levitsky H. Role of bone marrow-derived cells in presenting MHC class I-restricted tumor antigens. Science. 1994;264(5161):961–965. doi: 10.1126/science.7513904. [DOI] [PubMed] [Google Scholar]
- 9.Sotomayor EM, Borrello I, Rattis FM, et al. Cross-presentation of tumor antigens by bone marrow-derived antigen-presenting cells is the dominant mechanism in the induction of T-cell tolerance during B-cell lymphoma progression. Blood. 2001;98(4):1070–1077. doi: 10.1182/blood.v98.4.1070. [DOI] [PubMed] [Google Scholar]
- 10.Gabrilovich D, Pisarev V. Tumor escape from immune response: mechanisms and targets of activity. Curr Drug Targets. 2003;4(7):525–536. doi: 10.2174/1389450033490849. [DOI] [PubMed] [Google Scholar]
- 11*.Steinman RM, Inaba K, Turley S, Pierre P, Mellman I. Antigen capture, processing, and presentation by dendritic cells: recent cell biological studies. Hum Immunol. 1999;60(7):562–567. doi: 10.1016/s0198-8859(99)00030-0. Antigen uptake, processing and presentation by DCs are amenable to cell biological approaches. This likely explains the phenomenon of cross priming in the setting of transplantation and other clinical states. [DOI] [PubMed] [Google Scholar]
- 12.Nouri-Shirazi M, Banchereau J, Bell D, et al. Dendritic cells capture killed tumor cells and present their antigens to elicit tumor-specific immune responses. J Immunol. 2000;165(7):3797–3803. doi: 10.4049/jimmunol.165.7.3797. [DOI] [PubMed] [Google Scholar]
- 13.Restifo NP. Building better vaccines: how apoptotic cell death can induce inflammation and activate innate and adaptive immunity. Curr Opin Immunol. 2000;12(5):597–603. doi: 10.1016/s0952-7915(00)00148-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Steinman RM, Turley S, Mellman I, Inaba K. The induction of tolerance by dendritic cells that have captured apoptotic cells. J Exp Med. 2000;191(3):411–416. doi: 10.1084/jem.191.3.411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Basu S, Binder RJ, Suto R, Anderson KM, Srivastava PK. Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-kappa B pathway. Int Immunol. 2000;12(11):1539–1546. doi: 10.1093/intimm/12.11.1539. [DOI] [PubMed] [Google Scholar]
- 16.Maity A, McKenna WG, Muschel RJ. The molecular basis for cell cycle delays following ionizing radiation: a review. Radiother Oncol. 1994;31(1):1–13. doi: 10.1016/0167-8140(94)90408-1. [DOI] [PubMed] [Google Scholar]
- 17.Mashiba H, Matsunaga K, Hata K. Enhancement of radiosensitizing effect of the nitroimidazole derivative RK28 on the proliferation of MethA tumor cells in combined use with diethyldithiocarbamate. Life Sci. 1991;49(19):1419–1425. doi: 10.1016/0024-3205(91)90394-q. [DOI] [PubMed] [Google Scholar]
- 18.Candido KA, Shimizu K, McLaughlin JC, et al. Local administration of dendritic cells inhibits established breast tumor growth: implications for apoptosis-inducing agents. Cancer Res. 2001;61(1):228–236. [PubMed] [Google Scholar]
- 19**.Nikitina EY, Gabrilovich DI. Combination of gamma-irradiation and dendritic cell administration induces a potent antitumor response in tumor-bearing mice: approach to treatment of advanced stage cancer. Int J Cancer. 2001;94(6):825–833. doi: 10.1002/1097-0215(20011215)94:6<825::aid-ijc1545>3.0.co;2-5. The combination of gamma-irradiation and DC administration, but not each of the treatments alone resulted in a significant increase in T cell response to the specific, but not to the control peptides and in a dramatic antitumor effect. [DOI] [PubMed] [Google Scholar]
- 20**.Teitz-Tennenbaum S, Li Q, Rynkiewicz S, et al. Radiotherapy potentiates the therapeutic efficacy of intratumoral dendritic cell administration. Cancer Res. 2003;63(23):8466–8475. Intra-tumoral DC administration combined with RT induces a potent local and systemic antitumor response in tumor-bearing mice. This novel regimen may be beneficial in the treatment of human cancers. [PubMed] [Google Scholar]
- 21.Tong Y, Song W, Crystal RG. Combined intratumoral injection of bone marrow-derived dendritic cells and systemic chemotherapy to treat pre-existing murine tumors. Cancer Res. 2001;61(20):7530–7535. [PubMed] [Google Scholar]
- 22.Yu B, Kusmartsev S, Cheng F, et al. Effective combination of chemotherapy and dendritic cell administration for the treatment of advanced-stage experimental breast cancer. Clin Cancer Res. 2003;9(1):285–294. [PubMed] [Google Scholar]
- 23.Song W, Levy R. Therapeutic vaccination against murine lymphoma by intratumoral injection of naive dendritic cells. Cancer Res. 2005;65(13):5958–5964. doi: 10.1158/0008-5472.CAN-05-0406. [DOI] [PubMed] [Google Scholar]
- 24*.Keilholz U, Weber J, Finke JH, et al. Immunologic monitoring of cancer vaccine therapy: results of a workshop sponsored by the Society for Biological Therapy. J Immunother. 2002;25(2):97–138. doi: 10.1097/00002371-200203000-00001. Focus on various immune monitoring tools including assays of T-cell function, serologic and proliferation assays to assess B cell and T helper cell activity, and enzyme-linked immunospot assay, tetramer, cytokine flow cytometry, and reverse transcription polymerase chain reaction assays of T-cell immunity. [DOI] [PubMed] [Google Scholar]
- 25.Zhao J, Tenev T, Martins LM, Downward J, Lemoine NR. The ubiquitin-proteasome pathway regulates survivin degradation in a cell cycle-dependent manner. J Cell Sci. 2000;113(Pt 23):4363–4371. doi: 10.1242/jcs.113.23.4363. [DOI] [PubMed] [Google Scholar]
- 26.Rohayem J, Diestelkoetter P, Weigle B, et al. Antibody response to the tumor-associated inhibitor of apoptosis protein survivin in cancer patients. Cancer Res. 2000;60(7):1815–1817. [PubMed] [Google Scholar]
- 27.Andersen MH, Pedersen LO, Becker JC, Straten PT. Identification of a cytotoxic T lymphocyte response to the apoptosis inhibitor protein survivin in cancer patients. Cancer Res. 2001;61(3):869–872. [PubMed] [Google Scholar]
- 28.Schmitz M, Diestelkoetter P, Weigle B, et al. Generation of survivin-specific CD8+ T effector cells by dendritic cells pulsed with protein or selected peptides. Cancer Res. 2000;60(17):4845–4849. [PubMed] [Google Scholar]
- 29*.Pisarev V, Yu B, Salup R, Sherman S, Altieri DC, Gabrilovich DI. Full-length dominant-negative survivin for cancer immunotherapy. Clin Cancer Res. 2003;9(17):6523–6533. DCs transduced with dominant-negative survivin induce potent survivin-specific CTL responses able to recognize and kill tumor cells providing a rationale for immunotherapy clinical trials. [PubMed] [Google Scholar]
- 30.Kato J, Kuwabara Y, Mitani M, et al. Expression of survivin in esophageal cancer: correlation with the prognosis and response to chemotherapy. Int J Cancer. 2001;95(2):92–95. doi: 10.1002/1097-0215(20010320)95:2<92::aid-ijc1016>3.0.co;2-9. [DOI] [PubMed] [Google Scholar]
- 31.Kawasaki H, Toyoda M, Shinohara H, et al. Expression of survivin correlates with apoptosis, proliferation, and angiogenesis during human colorectal tumorigenesis. Cancer. 2001;91(11):2026–2032. doi: 10.1002/1097-0142(20010601)91:11<2026::aid-cncr1228>3.0.co;2-e. [DOI] [PubMed] [Google Scholar]
- 32.Miller M, Smith D, Windsor A, Kessling A. Survivin gene expression and prognosis in recurrent colorectal cancer. Gut. 2001;48(1):137–138. doi: 10.1136/gut.48.1.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Grossman D, McNiff JM, Li F, Altieri DC. Expression and targeting of the apoptosis inhibitor, survivin, in human melanoma. J Invest Dermatol. 1999;113(6):1076–1081. doi: 10.1046/j.1523-1747.1999.00776.x. [DOI] [PubMed] [Google Scholar]
- 34.Ito T, Shiraki K, Sugimoto K, et al. Survivin promotes cell proliferation in human hepatocellular carcinoma. Hepatology. 2000;31(5):1080–1085. doi: 10.1053/he.2000.6496. [DOI] [PubMed] [Google Scholar]
- 35.Smith SD, Wheeler MA, Plescia J, Colberg JW, Weiss RM, Altieri DC. Urine detection of survivin and diagnosis of bladder cancer. JAMA. 2001;285(3):324–328. doi: 10.1001/jama.285.3.324. [DOI] [PubMed] [Google Scholar]
- 36.Tanaka K, Iwamoto S, Gon G, Nohara T, Iwamoto M, Tanigawa N. Expression of survivin and its relationship to loss of apoptosis in breast carcinomas. Clin Cancer Res. 2000;6(1):127–134. [PubMed] [Google Scholar]
- 37.Moriai R, Asanuma K, Kobayashi D, et al. Quantitative analysis of the anti-apoptotic gene survivin expression in malignant haematopoietic cells. Anticancer Res. 2001;21(1B):595–600. [PubMed] [Google Scholar]
- 38.Sarela AI, Scott N, Ramsdale J, Markham AF, Guillou PJ. Immunohistochemical detection of the anti-apoptosis protein, survivin, predicts survival after curative resection of stage II colorectal carcinomas. Ann Surg Oncol. 2001;8(4):305–310. doi: 10.1007/s10434-001-0305-0. [DOI] [PubMed] [Google Scholar]
- 39*.Kappler M, Kotzsch M, Bartel F, et al. Elevated expression level of survivin protein in soft-tissue sarcomas is a strong independent predictor of survival. Clin Cancer Res. 2003;9(3):1098–1104. An elevated survivin content in tumor tissue extracts has a significant and independent negative predictive value on the survival-rate of STS patients. [PubMed] [Google Scholar]
- 40.Wurl P, Kappler M, Meye A, et al. Co-expression of survivin and TERT and risk of tumour-related death in patients with soft-tissue sarcoma. Lancet. 2002;359(9310):943–945. doi: 10.1016/S0140-6736(02)07990-4. [DOI] [PubMed] [Google Scholar]
- 41.Taubert H, Heidenreich C, Holzhausen HJ, et al. Expression of survivin detected by immunohistochemistry in the cytoplasm and in the nucleus is associated with prognosis of leiomyosarcoma and synovial sarcoma patients. BMC Cancer. 2010;10:65. doi: 10.1186/1471-2407-10-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Finkelstein SE, Gabrilovich DI, Bui MM, et al. At the Confluence of Radiation Therapy and Immunotherapy: External Beam Radiation (EBRT) with Intratumoral Injection of Dendritic Cells as Neoadjuvant Treatment of High-risk Soft Tissue Sarcoma Patients. Int J Radiat Oncol. 2009;75(3):S65–S65. doi: 10.1016/j.ijrobp.2010.12.068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43**.Finkelstein SE, Iclozan C, Bui MM, et al. Combination of External Beam Radiotherapy (EBRT) with Intratumoral Injection of Dendritic Cells as Neo-Adjuvant Treatment of High-Risk Soft Tissue Sarcoma Patients. Int J Radiat Oncol Biol Phys. 2011 doi: 10.1016/j.ijrobp.2010.12.068. Combination of intratumoral DC administration with EBRT was safe and resulted in induction of antitumor immune responses. This therapy is promising and needs further testing in clinical trials design to assess clinical efficacy. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Iclozan CD, Cotter MJ, Ramakrishnan R, et al. Myeloid-derived Suppressor Cell and Regulatory T Cell Immune Monitoring of Soft Tissue Sarcoma Patients Undergoing Combination External Beam Radiation and Intratumoral Dendritic Cell Vaccination. Int J Radiat Oncol. 2010;78(3):S657–S658. [Google Scholar]
- 45.Noyes DR, Lenox BC, Bui MM, et al. Serum IL-R1a and IP-10 Cytokine/chemokine Profiling Differences in Immune Responders and Non-responders to Combination Radiation/Immunotherapy for Soft Tissue Sarcoma. Int J Radiat Oncol. 2010;78(3):S85–S86. [Google Scholar]
- 46.Weber J. Immune checkpoint proteins: a new therapeutic paradigm for cancer--preclinical background: CTLA-4 and PD-1 blockade. Semin Oncol. 2010 Oct;37(5):430–9. doi: 10.1053/j.seminoncol.2010.09.005. [DOI] [PubMed] [Google Scholar]
- 47.Munn DH. Indoleamine 2,3-dioxygenase, Tregs and cancer. Curr Med Chem. 2011;18(15):2240–6. doi: 10.2174/092986711795656045. [DOI] [PubMed] [Google Scholar]
