APP下载

Nonmuscle invasive bladder cancer: a primer on immunotherapy

2016-08-10MahirMarufSamBrancatoPiyushAgarwal

Cancer Biology & Medicine 2016年2期

Mahir Maruf, Sam J. Brancato, Piyush K. Agarwal



Nonmuscle invasive bladder cancer: a primer on immunotherapy

Mahir Maruf, Sam J. Brancato, Piyush K. Agarwal

Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA

ABSTRACTIntravesical Bacillus Calmette-Guérin (BCG) has long been the gold standard treatment of nonmuscle invasive bladder cancer. Recently, there has been an emergence of novel immunotherapeutic agents, which have shown promise in the treatment of urothelial cell carcinoma. These agents aim to augment, modify, or enhance the immune response. Such strategies include recombinant BCG, monoclonal antibodies, vaccines, gene therapy, and adoptive T-cell therapy. Here, we review the emerging immunotherapeutics in the treatment of nonmuscle invasive bladder cancer.

KEYWORDSUrothelial cell carcinoma; Bacillus Calmette-Guérin (BCG); immunotherapy; bladder cancer

Introduction

Bladder cancer is the 9thmost common malignancy in the world. Urothelial cell carcinoma is the most common histological subtype accounting for 90% of all cases1. There was an estimated 74,000 newly diagnosed cases in the United States last year and 16,000 deaths attributed to bladder cancer2. Intravesical Bacillus Calmette-Guérin (BCG)represents one of the largest success stories in immunotherapy. It has been shown to decrease recurrence rates and progression to muscle invasive disease in patients with carcinoma in situ (CIS) and superficial tumors3,4. Treatment with a six-week course of intravesical BCG following transurethral resection remains the gold standard for managing patients with high-grade nonmuscle invasive bladder cancer (NMIBC). This review encompasses the most recent advances in immunotherapeutic strategies for NMIBC (Table 1).

BCG

BCG is a live attenuated strain of Mycobacterium bovis,originally developed in 1921 as a tuberculosis vaccine. The exact mechanism by which intravesical BCG exerts its therapeutic effect is not well understood. It is known to induce a local inflammation within the bladder mucosa following instillation, characterized by an influx of inflammatory cells and production of inflammatory cytokines. This cascade is initiated by the internalization of BCG into the urothelial cells via attachment of its fibronectin attachment protein to fibronectin, an extracellular matrix protein (Figure 1)5. This leads to the influx of granulocytes,macrophages, natural killer (NK) cells, dendritic cells and lymphocytes6,7. Cytokines such as IL-1, IL-2, IL-6, IL-8,tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) have been identified in the urine following repeated BCG instillations8. In addition to this non-specific inflammatory response, a BCG-specific immune response also plays a role in its antitumor effect. Antigen presentation to T cells is thought to be essential to the BCG response, as studies have shown that CD4+ or CD8+ T cell depleted mice have impaired antitumor activity9.

Although the generation of a Th1 mediated immune response is needed for BCG to exert its antitumor effect,there is growing evidence that the innate immune response may also contribute to the antitumor activity of BCG therapy. NK cells, display cytotoxic activity independent of MHC antigen presentation. To determine the antitumor property of NK cells, NK-deficient beige mice with were orthotopically implanted with MB49 bladder tumors with increasing doses of tumor burden and compared to wildtype mice with corresponding implanted tumor volumes. At the same tumor dose, the beige mice demonstrated reduced survival compared to their wildtype counterparts10. Thecytotoxic effect of NK cells results from the release of direct cytotoxic agents as the expression of secreted or membrane bound cytokines, including TNF-related apoptosis-inducing ligand (TRAIL)11. In response to IFN-α stimulation, T cells,NK cells, dendritic cells and macrophages express TRAIL,inducing downstream caspase activity in tumor cells bybinding to a family of death receptors, DR4 and DR5,ultimately resulting in apoptosis12. Neutrophils also express TRAIL in response to direct recognition of BCG cell wall components on toll-like-receptor 2 (TLR2), and amplify TRAIL expression in response to IFN-α13. Neutrophil recruitment is an early response to BCG, preceding T cell involvement, and occurs due to the release of chemokines such as IL-8 by the bladder epithelium. Inflammatory cytokine and TRAIL expression are necessary for the antitumor property of BCG therapy.

Table 1 Summary of immunotherapeutic options and their respective targets

Figure 1 Immune cascade as a proposed mechanism of action following intravesical BCG therapy. Adapted from Askland et al.5Adv. Urol. 2012.

Despite the efficacy of intravesical BCG for NMIBC, 20%-50% of patients will recur following one induction course14,15. Of these patients, only 25%-45% will respond to a second treatment course16. Additionally, the use of BCG may be limited in some patients due to severe local side effects or systemic toxicity.

Recombinant BCG (rBCG)

To decrease toxicity and improve the efficacy of intravesical BCG, rBCG has been utilized as an alternate immunotherapy strategy. The advent of genetic engineering has allowed for recombinant BCG strains to express immunomodulatory exogenous genes. Alternatively, BCG subcomponents have been engineered to decrease the local toxicity of BCG while still producing a comparable immunogenic response. The two major classifications of rBCG strategies are Th1 cytokine-secreting rBCG and non-live BCG subcomponent rBCG.

Th1 cytokine-secreting rBCG

A Th1 immunologic response is essential for BCG to produce its antitumor effect17. This has led to genetically manipulating BCG strains to secrete Th1 cytokines as a strategy to enhance effective therapy. Such strategies include IL-2, IL-12, IL-18, IFN-α, and IFN-γ secreting rBCG. In vitro immune assays have shown that IL-2 augments the cytotoxicity of NK cells as well as monocytes18,19. IL-2 is also essential for generating an immune response in vivo. In patients treated with intravesical BCG, studies have shown IL-2 levels peaks in the urine within 2-6 hours following treatment20,21. IL-2 rBCG profoundly potentiates the immune response22. In deer immunized with either BCG or IL-2 rBCG, IL-2 secreting rBCG induces a strong IFN-γ: IL-4 ratio, which may skew the immune response towards a Th1 response23,24.

IL-12 has demonstrated antitumor activity in murine adenocarcinoma and sarcoma models25. This cytokine induces production of high levels of IFN-γ and TNF-α. IL-12 also enhances NK activity, in addition to the alloreactive lymphocyte and cytotoxic T-cell responses. The role of IL-12 is not only to generate a Th-1 response, but also to inhibit the progression of a Th-2 immune response26. A study by O’Donnell et al.27showed that the use of IL-12 in an orthotopic murine bladder cancer model resulted in a dose dependent response of tumor regression and long term survival. Unfortunately recombinant IL-12 monotherapy has failed to show significant antitumor activity in phase 1 clinical trials28.

IL-18 acts synergistically with BCG to induce a Th1 immune response. Luo et al.29showed that IL-18 secreting rBCG results in increased IFN-γ production and augmented macrophage cytotoxicity in bladder cancer cells. The increased levels of IFN-γ may allow use of rBCG IL-18 alone or in combination with lower and safer doses of BCG30.

INF-α has been studied in bladder cancer cell lines in vitro and has demonstrated ability to induce apoptosis via TRAIL expression31. In addition, IFN-α has also demonstrated in vivo inhibition of tumor growth and tumor vascularization when studied in murine bladders implanted with human bladder cancer cells32. A large phase 2 trial studied the efficacy of IFN-α in combination with intravesical BCG administration, showing a 59% and 45% recurrence free survival at 24 months in BCG naïve and BCG failed patients,respectively. It is unclear if this treatment option is superior to intravesical BCG monotherapy, and a phase 3 trial would be necessary to investigate this further33. Although no trials to date have studied the efficacy of IFN-α as a single agent in humans, in vitro studies have shown rBCG-IFN-α to enhance the Th1 IFN-γ immune response of human peripheral blood mononuclear cells (PBMC). Compared to BCG alone, rBCGIFN-α showed superior PBMC cytotoxicity against human bladder cell lines34-36.

As previously discussed, IFN-γ is essential for cellmediated immunity and its role in BCG therapy has been well described, showing an inhibitory effect on bladder cancer cells37. In vitro rBCG-IFN-γ upregulates major histocompatibility complex (MHC) -1 in murine bladder cell lines, leading to incremental therapeutic efficacy in orthotopic mice38.

A subunit from the bacteria Bordetella pertussis, S1PT, has also been genetically engineered for expression in rBCG. Spleen cells from rBCG-S1PT-vaccinated mice exhibited increased levels of IFN-γ and decreased levels of IL-4, leading to a dominant cellular immune response and a decreased humoral immune response39,40. In a mouse orthotopic tumor model, rBCG-S1PT therapy resulted in bladder tumor weight reduction and increased survival time compared to the control group40.

BCG subcomponent based

Using non-live immunologically active BCG subcomponents is an attractive alternative to using live attenuated BCG as a method for decreasing the toxicity associated with BCG41. In one study, 61 patients with carcinoma in situ were treated with intravesical mycobacterium cell wall extract (MCWE). A negative biopsy was seen in 62.5%, 49.3%, and 41.1% of patients at 12, 24, and 60 weeks following treatment respectively42. Though these results are promising, to date there has not been a study with results that compare BCG cell wall components to local chemotherapy. A phase 3 study investigating the intravesical EN3348, a mycobacterial cell wall-DNA complex, compared to mytomycin C, was terminated early due to lack of accrual (NCT01200992). One limitation of MCWE is related to stability in solution making it challenging to use in the clinical. To overcome the unfavorable biophysical properties, Miyazaki et al.43used octaarginine liposomes (R8 liposomes) as a vector for transporting BCG cell wall skeleton into the cytoplasm of murine bladder tumor cells. This led to inhibition of bladder carcinogenesis making it a prospective substitute for BCG for immunotherapy against NMIBC43.

Mycobacterium phlei cell wall nucleic acid complex (MCNA) has been postulated to exert antineoplastic activity by exhibiting immunotherapeutic effect as well as direct chemotherapeutic effect, and has such has been used in a trial as intravesical therapy to treat high grade bladder cancer44. Although the study concluded that MCNA therapy lowered the risk of progression and cystectomies, the FDA advisory panel recently voted against its Biologics Application License,citing the lack of study power and lack of control group45. PstS1 is a phosphate binding subunit of the Mycobacterium permease protein found on the cell membrane46,47. This protein subunit is a remarkably immunogenic antigen48. In vitro data shows PstS1 may be exploited as a potent immunostimulatory antigen, resulting in increased cytotoxicity, IFN-γ release, and proliferation of PBMCs. When applied to in vivo experiments, therapeutic effects were observed in mice treated with intravesical recombinant PstS1 for orthotopic bladder tumors, as mice treated with intravesical PstS1 showed significantly prolonged survival compared to PBS control49. Other protein subcomponents such as MPT-64, and Ag85B have also demonstrated antitumor potential50,51.

Monoclonal antibodies

Monoclonal antibodies targeting tumor associated antigens(TAAs) are another novel immunotherapy strategy that may yield auspicious results. One example of such a TAA is βhCG, which is elevated in the serum and urine of 30%-40% of bladder cancer patients. Elevated β-hCG may be associated with advanced disease and increased mortality52. CDX-1307 is a monoclonal antibody that targets β-hCG, which may have utility in the treatment of bladder cancer. The antibody consists of B11, a monoclonal antibody against the mannose receptors of antigen presenting cells (APCs), fused to β-hCG. Internalization of CDX-1307 and subsequent presentation of the β-hCG on APCs leads to both cellular and humoral responses via antigen presentation to CD4+ and CD8+ T cells. This suggests that CDX-1307 may provide therapeutic benefit in the subset of patients with β-hCG expressing bladder cancer53.

Checkpoint inhibitors are a novel approach to overcoming the cancer induced immune dysfunction that would otherwise eliminate cancer cells before they are clinically evident. Tumors maintain immune tolerance through a variety of mechanisms including maintaining protective cell types in the tumor microenvironment, producing soluble molecular factors, and expressing co-inhibitory receptors. These co-inhibitory receptors are commonly referred to as immune checkpoints, and lead to a reticence of tumor specific effector cells such as T cells. Thus, strategies to interrupt the immune check points have recently been developed to overcome tumor immune resistance.

Activation of T cells requires two major stimulatory signals. The first is provided by the recognition of MHCs on APCs by the T cell receptor (TCR). The second signal is provided by B7 stimulatory molecules on APCs and recognized by CD28 on the T cell. Expansion and activation of the T cell follows the recognition of both the stimulatory signals. Activated T cells express cytotoxic T-lymphocyteassociated antigen 4 (CTLA-4) to compete with CD28 costimulatory receptor for the secondary B7 activation signal provided by the APC, thus preventing excessive T cell proliferation. T regulatory cells within the tumor microenvironment can constitutively express CTLA-4 attenuate tumor specific T cell response54. Ipilimumab is a monoclonal antibody against CTLA-4, and a phase 3 study showed increased survival in patients with metastatic melanoma who were administered the antibody. Patients randomized to the control arm received glycoprotein 100 (gp100) and demonstrated overall survival of 6.4 months. Patients in the experimental arm received ipilimumab, with or without gp100, and demonstrated an overall survival of 10.0 months, a significant improvement compared to gp100 alone. Though these studies show exciting results for patientswith metastatic disease, a study by Liakou et al.55studied the use of ipilimumab in presurgical patients with bladder cancer. Six patients with localized bladder cancer were treated with ipilimumab and were found to have expression of inducible costimulatory (ICOS), a T cell specific surface molecule structurally similar to CD28, on peripheral blood CD4+ T cells. In vitro studies of the CD4+ICOShicells from the ipilimumab treated patients showed an increased effector T cell to regulatory T cell ratio, as well as an increased production on IFN-γ55. A phase 1 trial studied the safety of using ipilimumab in 12 presurgical patients with localized urothelial cancer and showed the treatment was tolerable in 11 patients, with grade 1-2 toxicities, rash and diarrhea, being the most common56. Observations of the phenotypic biomarkers and tolerability are important steps needed in order to illuminate the mechanism of action and safety profile of ipilimumab, and are paramount for determining the clinical outcomes and of this antibody in the immunotherapy of bladder cancer.

The interaction between programmed death 1 (PD-1) on T-cells and its ligands, PD-L1 (B7-H1; CD274) and PD-L2 (B7-DC; CD273), is an immune check point explored as another viable option for checkpoint inhibition immunotherapy. PD-1, like CTLA-4, is a transmembrane protein found mainly on T cells, which serves as a regulator of lymphocytic function. In vitro assays show that the engagement of PD-1 with PD-L1 results in inhibition of TCR activation57,58. Thus, expression of PD-L1 acts to maintain immune tolerance and prevent autoimmunity. Urothelial tumors, like other solid tumors, can take advantage of this by expressing PD-L1 in order to develop immune tolerance59,60. Expression of PD-L1 on bladder cancer cells was associated with high grade tumors (odds ratio 2.4,95% CI 1.2-4.72,P=0.009) and was found to be extensively expressed in granulomas of recurrent tumors. Furthermore, PD-L1 acts as a marker for tumor progression, independent of tumor grade, and was associated with a higher frequency of postoperative recurrence and lower survival rate61,62. MPDL3280A is an anti-PD-L1 monoclonal antibody that inhibits the interaction between PD-1 and PD-L1. Administered systemically, the antibody has been studied in patients with metastatic urothelial bladder cancer in a recent phase 1 clinical trial63. In the evaluation of safety, 57% of 67 patients reported adverse events, while 4% of patients reported a grade 3 adverse event. No grade 4 or 5 adverse events were reported. The patients evaluated for efficacy had poor prognostic factors at baseline, including visceral metastasis in 50 patients (75%), while 48 patients (72%) had failed previous systemic therapy. After a 6 week follow up from treatment, objective response rates were 43% in patients with PD-L1 expression on tumor infiltrating immune cells by immunohistochemistry, representing remarkable clinical activity against metastatic bladder cancer. Other check point inhibitors such as B7-H3, B7-H4, LAG3,and TIM3 have been identified in basic studies, of which some are being studied in a clinical setting. However, to date,no data has been published demonstrating the clinical efficacy of monoclonal antibodies as checkpoint inhibitors for nonmuscle invasive bladder cancer45,64.

Vaccines

Vaccination is another form of emerging immunotherapy against NMIBC. Vaccines offer certain advantages over monoclonal antibody therapy, including decreased risk of tumor escape from targeting a single isotope and preventing tumor growth rather than requiring continual treatment of passive antibody treatment. Targets of vaccination include oncoproteins [e.g. HER2 and cancer testis antigens (CTAs)]as well as TAAs [e.g. carcinoembryonic antigen (CEA) and mucin-1 (MUC-1)]. A meta-analysis of nine studies found that HER2 expression in patients with bladder cancer was associated with higher histological grade, higher rates of lymph node metastasis, poor survival rates and poor disease free65. HER2 expression was positive in 27.8%-85.2% of bladder cancer making the oncoprotein a feasible vaccine target. A recently completed phase 2 trial investigated DN24-02, a form of autologous immunotherapy consisting leukapheresed APCs cultured in recombinant a HER2 antigen (NCT01353222). APC activation was observed in all of the first 30 patients who underwent three infusions in two week intervals, suggesting a boost in immunologic effect. The adverse effects were relatively minor, grades 1-266. A phase 1 clinical trial is currently ongoing at the National Institutes of Health investigating an autologous AdHER2/neu dendritic cell vaccine in patients with solid metastatic tumors and as an adjuvant in postcystectomy patients with HER2+ tumors (NCT01730118). In murine models, a recombinant adenoviral ErbB-2/neu vaccine demonstrated the ability to cure subcutaneous mammary tumors and pulmonary metastases67.

CTAs are a group of antigens that are normally expressed in male germ cells in the testis, but not in somatic tissue. Malignant cells may attain the ability to express CTAs by disruption of the regulated gene. As CTAs are immunogenic,they have become the target of developing antigen specific vaccines68. A study of tumor samples from 95 patients with high-grade urothelial carcinoma showed that 77% of tumorsexpressed at least one CTA with MAGE-A4 and MAGE-A3 being the most common CTA expressed69. As a result,MAGE-A4, along with the most immunogenic CTA in urothelial carcinoma, NY-ESO-1, are both currently being studied as targets for vaccine therapy in NMIBC70. For example, a study evaluated the safety and immunogenicity of an NY-ESO-1 vaccine in patients with urothelial carcinoma who had previously undergone cystectomy or nephroureterctomy. Enrolled patients were administered the NY-ESO-1 vaccine plus GM-CSF along with intradermal BCG. The study found 5 of 6 patients enrolled responded with anti-NY-ESO-1 specific antibodies and 1 of 6 patients exhibited a CD8+ response. All 6 patients displayed a CD4+ response71. Toxicities were limited to grade 1-2 in all patients. A phase 1 study evaluating the safety of an NY-ESO-1 vaccine with or without sirolimus in patients with solid tumors is currently being conducted and has completed accrual (NCT01522820). A recently completed phase 1 vaccine trial evaluated a CTA, MAGE-A3 (NCT01498172). Patients with NMIBC received intravesical BCG along with recMAGE-A3 immunization and adjuvant AS15 (recMAGEA3 + AS15 ASCI) in order to assess vaccine safety and assess vaccine specific T-cell response. At this time, no results are available. Another ongoing phase 2 trial is evaluating disease free survival and clinical efficacy of recMAGE-A3 + AS-15 in patients who underwent cystectomy for muscle invasive bladder cancer (NCT01435356).

The National Institutes of Health has an ongoing phase 2 trial assessing the clinical utility of PANVAC, a poxvirus based vaccine, in the treatment of NMIBC for patients who have failed at least one prior BCG course (NCT02015104). The PANVAC vaccine has shown efficacy in several other carcinomas expressing antigens such as MUC-1, CEA, and three T cell costimulatory molecules (B7.1, intracellular adhesion moldecule-1, and leukocyte function-associated antigen-3)72,73. Using immunohistochemistry, MUC-1 expression has been identified on 60%-70% of bladder cancers and increased expression is associated with higher grade tumors74-76. Similarly, CEA is expressed in approximately 76% of grade 4 tumors via immunohistochemistry77. As such, these immunogenic surface antigens make them plausible for use in vaccine immunotherapies.

Gene therapy

Gene-therapy refers to the genetic modification of normal or diseased cells to treat diseases, including cancers78. Advances in gene therapy have led to its application in pancreatic cancer79, malignant gliomas80, and lung cancer81. In bladder cancer, the use of gene therapy may be applied in the context of immunomodulation, direct oncolytic effect, and gene transfer. The entire application of gene therapy for the treatment of bladder cancer is beyond the scope of this review, and herein we will highlight some advances in gene therapy with respect to immunotherapy for superficial bladder cancer. A key component to gene therapy is the vector, or modality by which genes are delivered to the target cells. Vectors are employed to transduce a target cell with foreign DNA. Although numerous vectors have been described, the most commonly used vector in gene therapy is replication deficient adenovirus82. Adenovirus based gene therapy offer many advantages, including relative ease of producing, relative stability, and gene delivery capabilities,making it a commonly tested modality for gene-therapy in clinical trials83.

In a study by Chester et al.84, replication defective adenovirus with β-galactosidase marker gene was to transduce healthy human urothelial cells as well as neoplastic urothelial cells, demonstrating the ability to transduce human urothelial cells in in vitro and ex vivo models. Of the eight explant cultures of transitional cell carcinoma (TCC)were exposed to the adenovirus, four exhibited a near 100% transduction rate. In the same study, three fresh cold cup biopsy specimens of papillary TCC were exposed to the adenovirus. The superficial epithelial cells of the specimen demonstrated successful transduction. This study suggests the feasibility of using adenovirus as a vector for gene therapy for bladder cancer in human studies.

This principle has then been used in immunomodulating gene therapy. Transduced genes provide antitumor activity against NMIBC by sensitizing the cancer cells to the immune system. CD40L is protein found on Th1 helper cells and when bound to CD40 on APCs, is responsible for activation and antitumor activity of Th1 cells85. The use of the CD40L in an adenoviral vector (AdCD40L) has shown antitumor activity in a phase 1/2a trial for patients with high grade or Ta bladder cancer undergoing cystectomy86. Intravesical AdCD40L was administered and patient safety, along with tumor response, was monitored. Post treatment bladder biopsy samples from these patients had increased expression of CD40L on bladder epithelium as demonstrated by immunohistochemical staining. These epithelial samples were also infiltrated with CD4+ T cells. Antitumor activity was also remarkable as three of the five patients in the phase 1 trial demonstrated no tumor cells in the post treatment biopsy. Adverse events were limited to bladder spasms and mild bladder pain. As local AdCD40L therapy has shown tobe safe and immunogenic, it may play a role future bladder cancer therapy.

A phase 1 study by Dinney et al.87investigated the intravesical administration of recombinant adenovirus with an interferon-α2b gene (rAd-IFN-α) combined with an excipient, Syn3, in patients with BCG refractory NMIBC. The therapy was well tolerated as urgency, headache, fatigue, and nausea were the most commonly reported adverse events; no dose limiting toxicities were observed. Additionally,quantifiable levels of IFN-α were measured is all seventeen patients up to 10 days after the treatment. This therapy also demonstrated clinical activity as a complete response, as defined by a negative biopsy and no observable disease, was seen seven patients on 3-month follow up while five patients maintained complete response at 12 months. This study is the first to show evidence of safety and efficacy of intravesical recombinant adenoviral therapy for NMIBC.

Viral gene therapy is not without its obstacles. In 2003,systemic adenoviral gene therapy of a patient with ornithine transcarbamylase deficiency resulted in a fatal systemic inflammatory response syndrome, raising concerns regarding the safety of viral gene therapy88. However recent developments in intravesical delivery of viral vectors allows for local treatment of bladder cancer, thus avoiding complications of systemic administration84. Other obstacles include cellular susceptibility to viral infection. Adenovirus associated with the cell surface coxsackie-adenoviral receptor (CAR), resulting in intracellular incorporation of the virus. Neoplastic bladder epithelial cells are relatively CAR deficient, thus exhibiting increased adenoviral resistance89-91. Methods of increasing susceptibility, such as increasing cancer cell CAR expression92or modifying viral vectors93may pave a way for implementing viral gene therapy in clinical practice.

Adoptive T-cell therapy

Adoptive T-cell therapy, a form of passive immunotherapy,refers to the employment of autologous T-cell transfusions into tumor bearing patients in order to control tumor growth94. Tumors with lymphocytic infiltration have been associated with better prognosis; however, their small numbers are often not sufficient to correlate into a clinical effect95.

In vitro, tumor infiltrating lymphocytes (TILs) can be isolated and expanded without effects of inhibitory factors found in vivo. Researchers found that when these cells were isolated from a lymphodepleted patient with metastatic melanoma and then infused back into the patient, 21 (49%) of 43 patients experienced regression by RECIST criteria (Response Evaluation Criteria in Solid Tumors) criteria96. Grade 3-4 toxicities were observed with the lymphodepleting therapy, however no 3-4 toxicities were observed from administration of the autologous T-cells97.

Sherif et al.98infused autologous T lymphocytes from the metinel node of patients with advanced urothelial carcinoma T2N1 or higher to determine if this form of immunotherapy would be safe and technically feasible. Of the six patients in which the T lymphocytes were administered, no major adverse events were observed, warranting larger trials to determine long term clinical outcomes.

However, demonstrating clinical response in patients with bladder cancer presents certain challenges as epithelial cancers contain fewer mutations than melanoma and there is little data on endogenous mutation specific T-cell response99. Researchers at the National Cancer Institute have attempted to undertake these challenges as they have studied adoptive T cell therapy in a patient with metastatic cholangiocarcinoma100. Lung metastases were extracted from the patient for both whole genomic sequencing as well as T cell extraction. Sequencing of the tumor yielded 26 nonsynomymous mutations for each a minigene construct that was generated. Multiple minigenes were used to craft tandem minigene (TMG) constructs that were used for in vitro transcription, creating TMG RNAs. The RNAs were transfected into autologous APCs and cultured with the TILs extracted from the patient in order to determine which TILs would react to the transfected APCs. The patient was reinfused with the TILs. Results of the study suggest regression of the cancer and stabilization of the disease.

Adoptive T cell therapy still presents challenges, such as the need to perform an invasive extract procedure and difficulty growing TILs in some patients. To overcome such challenges, Clay et al.101used T cells from peripheral blood and transfected the T cells using a retrovirus to create genetically modified T cells with recombinant TCRs with an affinity for the TAA MART-1 in melanoma cells. Naïve T cells were successfully transduced to express a MART-1 specific TCR and demonstrated the ability to lyse melanoma cells in vitro.101A similar study was performed with autologous T cells transduced with TCR specific to NY-ESO-1 CT antigen. Patients with NY-ESO-1 expressing tumors were treated with modified T cells. A clinical response was observed in five of the eleven patients with metastatic melanoma and four of the six patients with synovial cell carcinoma102.

In addition to the benefit of avoiding technical difficulties,modified T cell therapy also provides the added benefit ofreducing immune escape due to MHC restrictions96. This has led to the development of genetically modified T cells with innate antitumor activity using chimeric antigen receptors (CARs). By grafting antigen binding domains of monoclonal antibodies to T cell signaling domains, CARs may bind directly to specific tumor antigens, bypassing stimulation from MHCs. Patients with relapsed acute lymphoblastic leukemia (ALL) were treated with CARs modified T cells specific for CD19, and a high remission rate was observed103. Given the exciting results modified T cell immunotherapy, it is not a far stretch that this may be an avenue for NMIBC treatment; however, research using urothelial models would be required to assess the utility and feasibility of this therapy.

Figure 2 Immunotherapy for superficial BCa, highlighted by the blue boxes, within the cycle of cancer-immune response. Adapted from Chen et al.104Immunity 2013.

Conclusions

Immunotherapy is an evolving field that offers promise in the treatment of NMIBC. Although BCG still remains the cornerstone treatment for high-grade NMIBC, the toxicity,limited efficacy in a subset of patients, and recurrence rates call for more effective treatment options. Recombinant BCG,monoclonal antibodies, vaccines, and adoptive immunotherapy are potential options aimed at addressing these shortcomings. The roles these therapies play in the cycle of cancer and immunity are displayed in Figure 2. Further research is necessary to bring immunotherapy further to the forefront of treating urothelial cancer. These treatment modalities may be combined with established treatments such as surgery, radiation, and chemotherapy to effectively treat bladder cancer.

Acknowledgement

This work was supported by the Intramural Research Program of the NIH, National Cancer Institute, Center for Cancer Research.

Conflict of interest statement

No potential conflicts of interest are disclosed.

References

1.Ploeg M, Aben KK, Kiemeney LA. The present and future burden of urinary bladder cancer in the world. World J Urol. 2009; 27: 289-93.

2.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015; 65: 5-29.

3.Herr HW, Laudone VP, Badalament RA, Oettgen HF, Sogani PC,Freedman BD, et al. Bacillus calmette-guerin therapy alters the progression of superficial bladder cancer. J Clin Oncol. 1988; 6: 1450-5.

4.Sylvester RJ, van der Meijden AP, Witjes JA, Kurth K. Bacillus calmette-guerin versus chemotherapy for the intravesical treatment of patients with carcinoma in situ of the bladder: A meta-analysis of the published results of randomized clinical trials. J Urol. 2005; 174: 86-91; discussion 91-2.

5.Askeland EJ, Newton MR, O'Donnell MA, Luo Y. Bladder cancer immunotherapy: BCG and beyond. Adv Urol. 2012; 2012: 181987.

6.Shintani Y, Sawada Y, Inagaki T, Kohjimoto Y, Uekado Y, Shinka T. Intravesical instillation therapy with bacillus calmette-guerin for superficial bladder cancer: Study of the mechanism of bacillus calmette-guerin immunotherapy. Int J Urol. 2007; 14: 140-6.

7.Bisiaux A, Thiounn N, Timsit MO, Eladaoui A, Chang HH, Mapes J, et al. Molecular analyte profiling of the early events and tissue conditioning following intravesical bacillus calmette-guerin therapy in patients with superficial bladder cancer. J Urol. 2009;181: 1571-80.

8.Jackson AM, Alexandroff AB, Kelly RW, Skibinska A,Esuvaranathan K, Prescott S, et al. Changes in urinary cytokines and soluble intercellular adhesion molecule-1(ICAM-1) in bladder cancer patients after Bacillus Calmette-Guerin (BCG)immunotherapy. Clin Exp Immunol. 1995; 99: 369-75.

9.Ratliff TL, Ritchey JK, Yuan JJ, Andriole GL, Catalona WJ. T-cell subsets required for intravesical bcg immunotherapy for bladder cancer. J Urol. 1993; 150: 1018-23.

10.Brandau S, Riemensberger J, Jacobsen M, Kemp D, Zhao W, Zhao X, et al. NK cells are essential for effective bcg immunotherapy. Int J Cancer. 2001; 92: 697-702.

11.Zamai L, Ahmad M, Bennett IM, Azzoni L, Alnemri ES, Perussia B. Natural killer (NK) cell-mediated cytotoxicity: Differential use of trail and fas ligand by immature and mature primary human nk cells. J Exp Med. 1998; 188: 2375-80.

12.Ludwig AT, Moore JM, Luo Y, Chen X, Saltsgaver NA, O'Donnell MA, et al. Tumor necrosis factor-related apoptosis-inducing ligand: A novel mechanism for bacillus calmette-guerin-induced antitumor activity. Cancer Res. 2004; 64: 3386-90.

13.Simons MP, O'Donnell MA, Griffith TS. Role of neutrophils in BCG immunotherapy for bladder cancer. Urol Oncol. 2008; 26: 341-5.

14.Cookson MS, Herr HW, Zhang ZF, Soloway S, Sogani PC, Fair WR. The treated natural history of high risk superficial bladder cancer: 15-year outcome. J Urol. 1997; 158: 62-7.

15.Lerner SP, Tangen CM, Sucharew H, Wood D, Crawford ED. Failure to achieve a complete response to induction bcg therapy is associated with increased risk of disease worsening and death in patients with high risk non-muscle invasive bladder cancer. Urol Oncol. 2009; 27: 155-9.

16.Haaff EO, Dresner SM, Ratliff TL, Catalona WJ. Two courses of intravesical bacillus calmette-guerin for transitional cell carcinoma of the bladder. T J Urol. 1986; 136: 820-4.

17.Wang Y, Yang M, Yu Q, Yu L, Shao S, Wang X. Recombinant Bacillus Calmette-Guerin in urothelial bladder cancer immunotherapy: Current strategies. Expert Rev Anticancer Ther. 2015; 15: 85-93.

18.Henney CS, Kuribayashi K, Kern DE, Gillis S. Interleukin-2 augments natural killer cell activity. Nature. 1981; 291: 335-8.

19.Malkovsky M, Loveland B, North M, Asherson GL, Gao L, Ward P, et al. Recombinant interleukin-2 directly augments the cytotoxicity of human monocytes. Nature. 1987; 325: 262-5.

20.De Jong WH, De Boer EC, Van der Meijden AP, Vegt P,Steerenberg PA, Debruyne FM, et al. Presence of interleukin-2 in urine of superficial bladder cancer patients after intravesical treatment with bacillus Calmette-Guérin. Cancer immunol immunother. 1990; 31: 182-6.

21.Ratliff TL, Haaff EO, Catalona WJ. Interleukin-2 production during intravesical bacille calmette-guerin therapy for bladder cancer. Clin immunol Immunopathol. 1986; 40: 375-9.

22.Murray PJ, Aldovini A, Young RA. Manipulation and potentiation of antimycobacterial immunity using recombinant bacille calmette-guerin strains that secrete cytokines. Proc Natl Acad Sci U S A. 1996; 93: 934-9.

23.Slobbe L, Lockhart E, O'Donnell MA, MacKintosh C, De Lisle G,Buchan G. An in vivo comparison of Bacillus Calmette-Guerin (BCG) and cytokine-secreting bcg vaccines. Immunology. 1999;96: 517-23.

24.Young S, O'Donnell M, Lockhart E, Buddle B, Slobbe L, Luo Y, et al. Manipulation of immune responses to mycobacterium bovis by vaccination with IL-2- and IL-18-secreting recombinant bacillus calmette guerin. Immunol Cell biol. 2002; 80: 209-15.

25.Nastala CL, Edington HD, McKinney TG, Tahara H, Nalesnik MA, Brunda MJ, et al. Recombinant IL-12 administration induces tumor regression in association with ifn-gamma production. J Immunol. 1994; 153: 1697-706.

26.Brunda MJ, Luistro L, Warrier RR, Wright RB, Hubbard BR,Murphy M, et al. Antitumor and antimetastatic activity of interleukin 12 against murine tumors. The Journal of experimental medicine. 1993; 178: 1223-30.

27.O'Donnell MA, Luo Y, Hunter SE, Chen X, Hayes LL, Clinton SK. Interleukin-12 immunotherapy of murine transitional cell carcinoma of the bladder: Dose dependent tumor eradication and generation of protective immunity. J Urol. 2004; 171: 1330-5.

28.Weiss GR, O'Donnell MA, Loughlin K, Zonno K, Laliberte RJ,Sherman ML. Phase 1 study of the intravesical administration of recombinant human interleukin-12 in patients with recurrent superficial transitional cell carcinoma of the bladder. J Immunother. 2003; 26: 343-8.

29.Luo Y, Yamada H, Chen X, Ryan AA, Evanoff DP, Triccas JA, et al. Recombinant Mycobacterium bovis bacillus Calmette-Guérin (BCG) expressing mouse IL-18 augments Th1 immunity and macrophage cytotoxicity. Clin Exp Immunol. 2004; 137: 24-34.

30.Luo Y, Chen XH, O'donnell MA. Role of Th1 and Th2 cytokines in BCG-induced IFN-gamma production: cytokine promotion and simulation of BCG effect. Cytokine. 2003; 21: 17-26.

31.Papageorgiou A, Dinney CP, Mcconkey DJ. Interferon-alpha induces TRAIL expression and cell death via an IRF-1-dependent mechanism in human bladder cancer cells. Cancer Biol Ther. 2007; 6: 872-9.

32.Slaton JW, Perrotte P, Inoue K, Dinney CP, Fidler IJ. Interferonalpha-mediated down-regulation of angiogenesis-related genes and therapy of bladder cancer are dependent on optimization of biological dose and schedule. Clin Cancer Res. 1999; 5: 2726-34.

33.Joudi FN, Smith BJ, O'Donnell MA. Final results from a nationalmulticenter phase 2 trial of combination bacillus Calmette-Guérin plus interferon alpha-2B for reducing recurrence of superficial bladder cancer. Urol Oncol. 2006; 24: 344-8.

34.Liu W, O'Donnell MA, Chen X, Han R, Luo Y. Recombinant bacillus Calmette-Guérin (BCG) expressing interferon-alpha 2b enhances human mononuclear cell cytotoxicity against bladder cancer cell lines in vitro. CancerImmunol Immunother.2009; 58: 1647-55.

35.Luo Y, Chen X, Han R, O'donnell MA. Recombinant Bacille Calmette-Guerin (BCG) expressing human interferon-alpha 2b demonstrates enhanced immunogenicity. Clin Exp Immunol. 2001; 123: 264-70.

36.Ding GQ, Yu YL, Shen ZJ, Zhou X-, Liao GD, Zhang YE. Antitumor effects of human interferon-alpha 2b secreted by recombinant bacillus Calmette-Guérin vaccine on bladder cancer cells. J Zhejiang Univ Sci B. 2012; 13: 335-41.

37.Hawkyard SJ, Jackson AM, James K, Prescott S, Smyth JF,Chisholm GD. The inhibitory effects of interferon gamma on the growth of bladder cancer cells. J Urol. 1992; 147: 1399-403.

38.Arnold J, De Boer EC, O'Donnell MA, Bohle A, Brandau S. Immunotherapy of experimental bladder cancer with recombinant bcg expressing interferon-gamma. J Immunother. 2004; 27: 116-23.

39.Nascimento IP, Dias WO, Mazzantini RP, Miyaji EN, Gamberini M, Quintilio W, et al. Recombinant mycobacterium bovis BCG expressing pertussis toxin subunit S1 induces protection against an intracerebral challenge with live bordetella pertussis in mice. Infect Immun. 2000; 68: 4877-83.

40.Andrade PM, Chade DC, Borra RC, Nascimento IP, Villanova FE,Leite LC, et al. The therapeutic potential of recombinant BCG expressing the antigen S1PT in the intravesical treatment of bladder cancer. Urol Oncol. 2010; 28: 520-5.

41.Zlotta AR, Van Vooren JP, Denis O, Drowart A, Daffe M, Lefevre P, et al. What are the immunologically active components of bacille calmette-guerin in therapy of superficial bladder cancer?Int J Cancer. 2000; 87: 844-52.

42.Morales A, Chin JL, Ramsey EW. Mycobacterial cell wall extract for treatment of carcinoma in situ of the bladder. J Urol. 2001;166: 1633-7; discussion 1637-8.

43.Miyazaki J, Nishiyama H, Yano I, Nakaya A, Kohama H, Kawai K,et al. The therapeutic effects of R8-liposome-BCG-CWS on BBN-induced rat urinary bladder carcinoma. Anticancer Res. 2011; 31: 2065-71.

44.Morales A, Herr H, Steinberg G, Given R, Cohen Z, Amrhein J, et al. Efficacy and safety of mcna in patients with nonmuscle invasive bladder cancer at high risk for recurrence and progression after failed treatment with bacillus Calmette-Guérin. J Urol. 2015; 193: 1135-43.

45.Wu Y, Enting D, Rudman S, Chowdhury S. Immunotherapy for urothelial cancer: from BCG to checkpoint inhibitors and beyond. Expert Rev Anticancer Ther. 2015; 15: 509-23.

46.Andersen AB, Hansen EB. Structure and mapping of antigenic domains of protein antigen b, a 38,000-molecular-weight protein of mycobacterium tuberculosis. Infect Immun. 1989; 57: 2481-8.

47.Braibant M, Lefèvre P, De Wit L, Peirs P, Ooms J, Huygen K, et al. A mycobacterium tuberculosis gene cluster encoding proteins of a phosphate transporter homologous to the escherichia coli Pst system. Gene. 1996; 176: 171-6.

48.Shi L, North R, Gennaro ML. Effect of growth state on transcription levels of genes encoding major secreted antigens of mycobacterium tuberculosis in the mouse lung. Infect Immun. 2004; 72: 2420-4.

49.Sanger C, Busche A, Bentien G, Spallek R, Jonas F, Bohle A, et al. Immunodominant psts1 antigen of mycobacterium tuberculosis is a potent biological response modifier for the treatment of bladder cancer. BMC Cancer. 2004; 4: 86.

50.Yu DS, Lee CF, Chang SY. Immunotherapy for orthotopic murine bladder cancer using bacillus Calmette-Guerin recombinant protein Mpt-64. J Urol. 2007; 177: 738-42.

51.Begnini KR, Rizzi C, Campos VF, Borsuk S, Schultze E, Yurgel VC, et al. Auxotrophic recombinant Mycobacterium bovis BCG overexpressing Ag85B enhances cytotoxicity on superficial bladder cancer cells in vitro. Appl Microbiol Biotechnol. 2013; 97: 1543-52.

52.Iles RK. Ectopic hCGbeta expression by epithelial cancer: Malignant behaviour, metastasis and inhibition of tumor cell apoptosis. Mol Cell Endocrinol. 2007; 260-2: 264-70.

53.Morse MA, Bradley DA, Keler TA, Green JA, Davis TA. CDX-1307: a novel vaccine under study as treatment for muscleinvasive bladder cancer. Expert Rev Vaccines. 2011; 10: 733-42.

54.Rothschild SI, Thommen DS, Moersig W, Muller P, Zippelius A. Cancer immunology - development of novel anti-cancer therapies. Swiss Med Wkly. 2015; 145: w14066.

55.Liakou CI, Kamat A, Tang DN, Chen H, Sun J, Troncoso P, et al. CTLA-4 blockade increases IFNgamma-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc Natl Acad Sci U S A. 2008; 105: 14987-92.

56.Carthon BC, Wolchok JD, Yuan J, Kamat A, Ng Tang DS, Sun J, et al. Preoperative CTLA-4 blockade: tolerability and immune monitoring in the setting of a presurgical clinical trial. Clin Cancer Res. 2010; 16: 2861-71.

57.Eppihimer MJ, Gunn J, Freeman GJ, Greenfield EA, Chernova T,Erickson J, et al. Expression and regulation of the PD-L1 immunoinhibitory molecule on microvascular endothelial cells. Microcirculation. 2002; 9: 133-45.

58.Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008; 26: 677-704.

59.Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol. 2008; 8: 467-77.

60.Hafez N, Petrylak DP. Could PD-L1 prove to be an effective therapeutic target for bladder cancer? Immunotherapy. 2015; 7: 1-2.

61.Inman BA, Sebo TJ, Frigola X, Dong H, Bergstralh EJ, Frank I, et al. PD-L1(B7-H1) expression by urothelial carcinoma of the bladder and BCG-induced granulomata: associations withlocalized stage progression. Cancer. 2007; 109: 1499-505.

62.Nakanishi J, Wada Y, Matsumoto K, Azuma M, Kikuchi K, Ueda S. Overexpression of B7-H1(PD-L1) significantly associates with tumor grade and postoperative prognosis in human urothelial cancers. Cancer Immunol Immunother. 2007; 56: 1173-82.

63.Powles T, Eder JP, Fine GD, Braiteh FS, Loriot Y, Cruz C, et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature. 2014; 515: 558-62.

64.Carosella ED, Ploussard G, Lemaoult JA. A systematic review of immunotherapy in urologic cancer: evolving roles for targeting of CTLA-4, PD-1/PD-L1, and HLA-G. Eur Urol. 2015; 68: 267-79.

65.Zhao J, Xu W, Zhang Z, Song R, Zeng S, Sun Y, et al. Prognostic role of HER2 expression in bladder cancer: a systematic review and meta-analysis. Int Urol Nephrol. 2015; 47: 87-94.

66.Bajorin DF, Sharma P, Gomella LG, Plimack ER, O'Donnell PH,Hoffman-Censits JH, et al. NeuACT, a phase 2, randomized,open-label trial of DN24-02: Updated analysis of HER2 expression, immune responses, product parameters, and safety in patients with surgically resected HER2+ urothelial cancer. J Clin Oncol. 2014; 32(suppl 4): abstr 296.

67.Park JM, Terabe M, Steel JC, Forni G, Sakai Y, Morris JC, et al. Therapy of advanced established murine breast cancer with a recombinant adenoviral ErbB-2/neu vaccine. Cancer Res. 2008;68: 1979-87.

68.Scanlan MJ, Simpson AJ, Old LJ. The cancer/testis genes: review,standardization, and commentary. Cancer Immun. 2004; 4: 1.

69.Sharma P, Shen Y, Wen S, Bajorin DF, Reuter VE, Old LJ, et al. Cancer-testis antigens: expression and correlation with survival in human urothelial carcinoma. Clin Cancer Res. 2006; 12: 5442-7.

70.Sharma P, Gnjatic S, Jungbluth AA, Williamson B, Herr H,Stockert E, et al. Frequency of NY-ESO-1 and LAGE-1 expression in bladder cancer and evidence of a new NY-ESO-1 T-cell epitope in a patient with bladder cancer. Cancer immun. 2003; 3: 19.

71.Sharma P, Bajorin DF, Jungbluth AA, Herr H, Old LJ, Gnjatic S. Immune responses detected in urothelial carcinoma patients after vaccination with NY-ESO-1 protein plus BCG and GM-CSF. J Immunother. 2008; 31: 849-57.

72.Madan RA, Arlen PM, Gulley JL. PANVAC-VF: poxviral-based vaccine therapy targeting CEA and MUC1 in carcinoma. Expert Opin Biol Ther. 2007; 7: 543-54.

73.Morse MA, Niedzwiecki D, Marshall JL, Chang DZ, Aklilu M,Crocenzi TS, et al. A randomized phase 2 study of immunization with dendritic cells modified with poxvectors encoding CEA and MUC1 compared with the same poxvectors plus GM-CSF for resected metastatic colorectal cancer. Ann Surg. 2013; 258: 879-86.

74.Goldenberg DM, Sharkey RM, Primus FJ. Immunocytochemical detection of carcinoembryonic antigen in conventional histopathology specimens. Cancer. 1978; 42: 1546-53.

75.Walsh MD, Hohn BG, Thong W, Devine PL, Gardiner RA,Samaratunga ML, et al. Mucin expression by transitional cell carcinomas of the bladder. Br J Urol. 1994; 73: 256-62.

76.Cardillo MR, Castagna G, Memeo L, De Bernardinis E, Di Silverio F. Epidermal growth factor receptor, MUC-1 and MUC-2 in bladder cancer. J Exp Clin Cancer Res. 2000; 19: 225-33.

77.Jautzke G, Altenaehr E. Immunohistochemical demonstration of carcinoembryonic antigen (CEA) and its correlation with grading and staging on tissue sections of urinary bladder carcinomas. Cancer. 1982; 50: 2052-6.

78.Mulligan RC. The basic science of gene therapy. Science. 1993;260: 926-32.

79.Tseng JF, Mulligan RC. Gene therapy for pancreatic cancer. Surg Oncol Clin N Am. 2002; 11: 537-69.

80.Prasad G, Wang H, Hill DL, Zhang R. Recent advances in experimental molecular therapeutics for malignant gliomas. Curr Med Chem Anticancer Agents. 2004; 4: 347-61.

81.Vattemi E, Claudio PP. Gene therapy for lung cancer: Practice and promise. Ann Ital Chir.2004; 75: 279-89.

82.Rosser CJ, Benedict WF, Dinney CP. Gene therapy for superficial bladder cancer. Expert Rev Anticancer Ther. 2001; 1: 531-9.

83.Kaplan JM. Adenovirus-based cancer gene therapy. Curr Gene Ther. 2005; 5: 595-605.

84.Chester JD, Kennedy W, Hall GD, Selby PJ, Knowles MA. Adenovirus-mediated gene therapy for bladder cancer: efficient gene delivery to normal and malignant human urothelial cells in vitro and ex vivo. Gene Ther. 2003; 10: 172-9.

85.Kikuchi T, Crystal RG. Anti-tumor immunity induced by in vivo adenovirus vector-mediated expression of CD40 ligand in tumor cells. Hum Gene Ther. 1999; 10: 1375-87.

86.Malmström PU, Loskog AS, Lindqvist CA, Mangsbo SM,Fransson M, Wanders A, et al. AdCD40L immunogene therapy for bladder carcinoma--the first phase 1/2a trial. Clin Cancer Res. 2010; 16: 3279-87.

87.Dinney CP, Fisher MB, Navai N, O'donnell MA, Cutler D,Abraham A, et al. Phase I trial of intravesical recombinant adenovirus mediated interferon-α2b formulated in Syn3 for Bacillus Calmette-Guérin failures in nonmuscle invasive bladder cancer. J Urol. 2013; 190: 850-6.

88.Raper SE, Chirmule N, Lee FS, Wivel N, Bagg A, Gao GP, et al. Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Mol Genet Metab. 2003; 80: 148-58.

89.Li YM, Pong RC, Bergelson JM, Hall MC, Sagalowsky AI, Tseng CP, et al. Loss of adenoviral receptor expression in human bladder cancer cells: A potential impact on the efficacy of gene therapy. Cancer Res. 1999; 59: 325-30.

90.Loskog A, Hedlund T, Wester K, De La Torre M, Philipson L,Malmström PU, et al. Human urinary bladder carcinomas Express adenovirus attachment and internalization receptors. Gene Ther. 2002; 9: 547-53.

91.Buscarini M, Quek ML, Gilliam-Hegarich S, Kasahara N, Bochner B. Adenoviral receptor expression of normal bladder and transitional cell carcinoma of the bladder. Urol Int. 2007; 78: 160-6.

92.Segura-Pacheco B, Avalos B, Rangel E, Velazquez D, Cabrera G. HDAC inhibitor valproic acid upregulates CAR in vitro and in vivo. Genet Vaccines Ther. 2007; 5: 10.

93.Hiwasa K, Nagaya H, Terao S, Acharya B, Hamada K, Mizuguchi H, et al. Improved gene transfer into bladder cancer cells using adenovirus vector containing RGD motif. Anticancer Res. 2012;32: 3137-40.

94.June CH. Adoptive t cell therapy for cancer in the clinic. J Clin Invest 2007; 117: 1466-76.

95.Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015; 348: 62-8.

96.Duong CP, Yong CS, Kershaw MH, Slaney CY, Darcy PK. Cancer immunotherapy utilizing gene-modified t cells:From the bench to the clinic. Mol Immunol. 2015; 67: 46-57.

97.Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL,Restifo NP, et al. Adoptive cell transfer therapy following nonmyeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol. 2005; 23: 2346-57.

98.Sherif A, Hasan MN, Marits P, Karlsson M, Winqvist O, Thorn M. Feasibility of T-cell-based adoptive immunotherapy in the first 12 patients with advanced urothelial urinary bladder cancer. Preliminary data on a new immunologic treatment based on the sentinel node concept. Eur Urol. 2010; 58: 105-11.

99.Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA,Kinzler KW. Cancer genome landscapes. Science. 2013; 339: 1546-58.

100.Tran E, Turcotte S, Gros A, Robbins PF, Dudley ME, Wunderlich JR, et al. Cancer immunotherapy based on Mutation-Specific CD4+T cells in a patient with epithelial cancer. Science. 2014; 344: 641-5.

101.Clay TM, Custer MC, Sachs J, Hwu P, Rosenberg SA, Nishimura MI. Efficient transfer of a tumor antigen-reactive TCR to human peripheral blood lymphocytes confers anti-tumor reactivity. J Immunol. 1999; 163: 507-13.

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

103.Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014; 371: 1507-17.

104.Chen DS, Mellman I. Oncology meets immunology: the cancerimmunity cycle. Immunity. 2013; 39: 1-10.

Cite this article as: Maruf M, Brancato SJ, Agarwal PK. Nonmuscle invasive bladder cancer: a primer on immunotherapy. Cancer Biol Med. 2016; 13: 194-205. doi: 10.20892/j.issn.2095-3941.2016.0020

Correspondence to: Piyush K. Agarwal

E-mail: piyush.agarwal@nih.gov

Received February 27, 2016; accepted March 31, 2016. Available at www.cancerbiomed.org

Copyright © 2016 by Cancer Biology & Medicine