Oral Oncology xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Oral Oncology journal homepage: www.elsevier.com/locate/oraloncology

Immunotherapy for head and neck squamous cell carcinoma Qiao Li, Mark E.P. Prince, Jeffrey S. Moyer ⇑ University of Michigan Comprehensive Cancer Center, 1500 E Medical Center Dr., Ann Arbor, MI 48109, United States

a r t i c l e

i n f o

Article history: Received 2 November 2014 Received in revised form 24 November 2014 Accepted 10 December 2014 Available online xxxx Keywords: Monoclonal antibody therapy Cytokine therapy Cancer vaccines T cell-based therapy Cancer stem cells

s u m m a r y Objectives: To review the current state of immunotherapy of head and neck squamous cell carcinoma. Materials and Methods: Review of the literature with emphasis on clinical trial data. Results: Patients with head and neck squamous cell carcinoma (HNSCC) have long been known to be immunosuppressed. This impairment of the immune system is believed, at least in part, to underlie the poor outcomes in this patient population. Modulating the immune system to improve cancer outcomes is an attractive concept in this difficult to treat population. Conclusion: New studies have started to unravel the mechanisms of immunosuppression and new therapies are being developed to exploit this new information. Ó 2015 Elsevier Ltd. All rights reserved.

Immune defects in HNSCC and immunotherapy Many human malignancies, including head and neck squamous cell carcinoma (HNSCC) are associated with quantitative and qualitative deficiencies in the immune system. Patients with HNSCC have lower absolute lymphocyte counts [1], higher immunosuppressive regulatory T (Treg) cells [2,3], and higher tumorassociated macrophages [4] when compared to healthy controls. These immune defects also include down-regulation of tumor leukocyte antigen expression [5] and spontaneous apoptosis of cytotoxic T-lymphocytes (CTLs) [6] along with the direct inhibition of immune defenses by the tumor secretion of vascular endothelial growth factor (VEGF) [7], prostaglandin E2 [8,9], transforming growth factor(TGF)-ß [10], and interleukin (IL)-10 [10]. Qualitative abnormalities also include impaired natural killer (NK) activity [11,12], impaired maturation of plasmacytoid dendritic cells (DCs) [13], and poor antigen-presenting function [14–17]. With an enhanced awareness of the immune defects present in HNSCC as well as much greater understanding of the basic mechanisms of the immune system in carcinogenesis, rationale therapeutic strategies are being developed to take advantage of this new knowledge. Five broad approaches are gaining popularity in the immunotherapy of HNSCC: (1) monoclonal antibody therapy; (2) cytokine therapy; (3) cancer vaccines; (4) T cell-based therapy, and (5) immunological targeting of HNSCC cancer stem cells.

⇑ Corresponding author at: University of Michigan Medical Center, 1500 E Medical Center Drive, TC 1904, Ann Arbor, MI 48109, United States. E-mail address: [email protected] (J.S. Moyer).

Conventional chemotherapy and radiation-conferred antitumor immune effects There is a greater appreciation for the role of the immune system in the mechanisms of tumor eradication with conventional treatment modalities. Conventional thinking is that cytotoxic chemotherapy and/or therapeutic external beam radiation act directly on the tumor. While certainly true in many instances, a growing body of preclinical and clinical data would suggest that the immune system plays a significant and meaningful role in tumor elimination with these standard treatment approaches. Initially introduced in 1953, the abscopal bystander effect is the regression of tumor outside of the radiation field [18]. Typically seen in melanoma, lymphoma, and renal-cell carcinoma, this effect is believed to be dependent on an intact immune system [19]. Several reports in humans have demonstrated distant tumor responses outside the radiation fields that were correlated with elevated tumor-specific antibodies and T-cell activation [20,21]. Taken together, these observations lend indirect support to the concept that the immune system is involved in chemoradiation. More direct support for the immune system’s involvement in chemoradiation comes from the danger signals released by dying cells. Apetoh et al. [22] describe a pathway in both mice and humans where dying tumor cells release a protein [high mobility-group box 1 (HMGB10)] that can activate the innate and adaptive immune system through Toll-like receptor 4 (TLR4) expressed on dendritic cells (DCs). In this study [22], patients with breast cancer who possess a TLR4 loss-of-function allele recur more quickly after chemoradiotherapy than those carrying the wild-type TLR4 allele.

http://dx.doi.org/10.1016/j.oraloncology.2014.12.005 1368-8375/Ó 2015 Elsevier Ltd. All rights reserved.

Please cite this article in press as: Li Q et al. Immunotherapy for head and neck squamous cell carcinoma. Oral Oncol (2015), http://dx.doi.org/10.1016/ j.oraloncology.2014.12.005

2

Q. Li et al. / Oral Oncology xxx (2015) xxx–xxx

High dose, or maximally tolerated doses (MTD), chemotherapy is well known to be immunosuppressive through direct effects on the immune system. However, low dose or metronomic chemotherapy may be more immunostimulating than immunosuppressive [23]. The immunostimulatory effects of metronomic chemotherapy include immunogenic cell death by increased antigen presentation [24–26], depletion of immunosuppressive Treg [27–30] and myeloid-derived suppressor cells (MDSC) [31], and enhancement of cytotoxic effector cells [32,33]. The effects of cytotoxic chemotherapy and radiation alone on the immune system make this an attractive approach to use in conjunction with immunotherapeutic strategies. Modulating the tumor microenvironment or the host immune system to be more receptive to systemic immunotherapy has strong support in preclinical models [34–37]. The use of chemotherapy and/or radiation along with immunotherapy in humans will be discussed below. Monoclonal antibody therapies Monoclonal antibody therapy is currently the most widely used form of immunotherapy in cancer patients [38]. Monoclonal antibodies directed against CD20 and HER-2 are standard of care in hematopoietic malignancies and breast cancer, respectively [38]. Epidermal growth factor receptor Monocloncal antibodies targeting the epidermal growth factor receptor (EGFR) are the most common immunotherapy in HNSCC [39]. EGFR leads to proliferation, survival, and metastasis in HNSCC and is mediated by oncogenic signaling along the RAS/MAPK and PI3K/Akt pathways [40]. While 90% of HNSCCs overexpress EGFR [41], high levels of EGFR do not predict response to cetuximab [42,43] and EGFR pathway inhibitors (intracellular tyrosine kinase) are not associated with meaningful clinical responses [44,45]. These observations argue for additional mechanisms of antitumor activity rather than just the inhibition of the signaling cascade with EGFR activation. Innate and adaptive immunity including antibody-dependent cellular cytotoxicity (ADCC) appear to have a significant role in the mechanism of action of cextuximab [46]. Cetuximab is the first and best studied monoclonal antibody against EGFR and is a chimeric IgG1 isotype containing a human constant region and a mouse variable region. Bonner et al. [47] studied 424 patients with HNSCC that were randomized to either radiation alone or radiation and cextuximab and found a significant increase in overall survival from 29 months to 49 months with the addition of cextuximab. Much of this survival advantage was seen in oropharyngeal patients where HPV status may have played a role in treatment efficacy. Nimotuzumab is also a human/mouse chimeric monoclonal antibody targeting EGFR and has been studied in several Phase II clinical trials of patients receiving definitive chemoradiation [48,49]. In both studies of advanced-stage HNSCC, the addition of nimotuzumab conferred a significant survival advantage. Panitumumab, a fully humanized monoclonal antibody specific to EGFR, has also been studied in HNSCC with less demonstrated efficacy. The SPECTRUM trial was a Phase 3 trial in 657 patients with recurrent or metastatic HNSCC comparing cisplatin and fluorouracil with and without panitumumab. The trial did not show improved overall survival but did demonstrate improved progression-free survival [50]. These finding are in contrast to the EXTREME study where overall survival was improved in patients who received cetuximab in addition to cisplatin or carboplatin and fluorouracil [51]. While one explanation for the difference between the studies may be study methodology, the fact that panitumumab is a fully humanized IgG2 isotype may be relevant.

Unlike cetuximab, panitumumab does not induce NK-DC cross priming [52]. It is unclear whether the different isotype of panitumumab, the lack of murine variable region, or other factors are responsible for the effect difference between panitumumab and cetuximab despite both binding to EGFR. However, one likely explanation is the difference in the immunogenicity (mouse versus human variable regions) between these different monoclonal antibodies. Vascularized endothelial growth factor Elevated pretreatment serum vascularized endothelial growth factor (VEGF) levels tend to indicate a more aggressive disease state and a poorer overall survival in advanced laryngeal carcinoma [53]. In a meta-analysis of 12 studies that examined VEGF levels in over 1000 HNSCC patients, tumors that overexpressed VEGF measured by immunohistochemistry were associated with 1.88 greater risk of death at 2 years [54]. Several studies have examined the use of bevacizumab in HNSCC. Bevacizumab is a recombinant humanized IgG1 monoclonal antibody that binds VEGF-A and was the first agent against this protein approved by the Food and Drug Administration. Fury et al. [55] undertook a phase 2 trial of bevacizumab with cisplatin and intensity-modulated radiation in advanced HNSCC that demonstrated a 2-year overall survival (OS) rate of 88% and progression-free survival (PFS) rate of 76%. Bevacizumab was also studied with standard radiation therapy and docetaxol with encouraging results. In 30 patients with over 3 years median follow-up, OS and PFS were 62% and 68%, respectively [56]. The combination of cetuximab and bevacizumab has also been investigated in recurrent or metastatic HNSCC. Forty-six patients were enrolled in this Phase 2 study that demonstrated a median PFS interval of 2.8 months and OS of 7.5 months in a patient population that was highly resistant to standard therapy (73% of patients recurred within 6 months of prior curative therapy) [57]. Despite the decrease in VEGF levels in patients treated with bevacizumab, the actual VEGF levels post-treatment have not been shown to be consistently associated with efficacy. Similar to cetuximab, bevacizumab has been shown, however, to have significant impacts on immunological parameters that are distinct from the VEGF pathways. In metastatic melanoma and ovarian cancer, treatment with bevacizumab is associated with significant increases in CD8+ effector cells [58,59] and a reduction in circulating T reg cells [58]. It would be reasonable to conclude that similar mechanisms may be present in HNSCC. Immune checkpoint blockade The immune response is a carefully orchestrated balance between potentiation and inhibition. While the lack of potentiation may be responsible for anergy and tumor progression, the lack of immune inhibition can result in autoimmunity and an unchecked immune response. In cancer immunotherapy, however, where immune suppressive mechanisms are elevated, pathways that limit the breaks on the immune system can be exploited. One pathway where this interaction is important in tumor formation is the stimulatory B7/CD28 pathway of antigen presenting cells and T cells and the inhibitory effects of CTLA4 and PD-1 on activated T cells that act as a break on this response. Monoclonal antibodies that block CTLA4 and PD-1 and release the breaks on the immune response have been used with success in several clinical trials. In 2010, ipilimumab became the first FDA-approved monoclonal antibody that blocks the inhibitory CTLA4 pathway. This approval was based on a randomized trial where ipilimumab alone was found to offer a survival benefit for patients with metastatic melanoma [60]. Long-term survival beyond 2 years was seen in

Please cite this article in press as: Li Q et al. Immunotherapy for head and neck squamous cell carcinoma. Oral Oncol (2015), http://dx.doi.org/10.1016/ j.oraloncology.2014.12.005

Q. Li et al. / Oral Oncology xxx (2015) xxx–xxx

18% of treated patients suggesting that this therapeutic approach could potentially have long-lasting immune effects after a relatively short course of treatment [61]. This approach would seem rationale in HNSCC given the fact that Tregs in the tumor microenvironment express high levels of CTLA4 [62,63]. A Phase 1b study of locally advanced HNSCC at the University of Pittsburgh School of Medicine examining ipilimumab with standard therapy (IMRT and cetuximab) is ongoing and has enrolled 3 patients as of January 2014 (NCT01935921) [40,46,64]. Whereas CTLA4 primarily limits effector T cell activation, the major role of PD1 is to limit T cell activity in the inflammatory immune response and to regulate autoimmunity [61]. PD1 is more broadly expressed than CTLA4 and is present on activated T cells, NK cells, B lymphocytes, monocytes, and DCs [61]. The ligand for PD1, PDL1, is found on the tumor surface of many solid tumor malignancies including HNSCC [65–69]. PDL1 is also found on myeloid cells in the tumor microenvironment [70–72]. Monoclonal antibodies to PD1 and PDL1 are currently being developed by various companies. In the first Phase 1 clinical trial of nivolumab, a fully humanized PD1 antibody (colon, renal, lung cancer, and melanoma), there were noted to be partial responses and a complete response that continued more than a year from the cessation of therapy [61,73]. Importantly, positive responses were correlated with increasing PDL1 expression on tumors [61]. Based on this early work, a multicenter, open-label, randomized study with pembrolizumab in patients with unresectable or metastatic melanoma showed significant benefit leading to accelerated approval by the FDA in September of 2014. Similarly, in a large Phase 3 study in metastatic melanoma of nivolumab compared to dacarbazine, nivolumab was associated with significant improvements in OS and PFS [74]. Seiwert et al. reported on toxicity data from pembrolizumab in patients with HNSCC and found that the medication is well tolerated in this patient population with few serious adverse events (NCT01848834) [75]. There are several trials with either pembrolizumab or nivolumab in HNSCC currently recruiting study participants. A Phase 3 study investigating nivolumab versus standard therapy in recurrent or metastatic HNSCC is ongoing (NCT02105636). Primary endpoints are PFS and OS with an estimated final data collection date in June 2016. A Phase 2 study examining pembrolizumab in recurrent or metastatic HNSCC after treatment with platinum-based and cetuximab therapy recently opened in October 2014 (NCT02255097). Primary outcomes to be evaluated are objective response rate (ORR) by RECIST. There are no definitive data yet available in HNSCC for CTLA4 and PD1 monoclonal antibody therapies. However the preclinical data and preliminary clinical data in other solid tumor malignancies are encouraging. CTLA4 or PD1 blockade will likely be more effective if combined with other immunologic modulating therapies, including standard treatment modalities, rather than used alone. These studies are an active area of interest for many investigators.

Cytokine therapy Cytokine-based immunotherapy has been used both locoregionally and systemically with limited success in HNSCC, albeit with significant toxicities in some patients. IL-2 is perhaps the most studied and has met with the most successes. The main function of the proinflammatory cytokine IL-2 is to promote antigen-specific clonal expansion of T cells as well as potentiate non-specific NK and LAK cell activity. Complete or partial responses have been reported with IL-2 therapy in HNSCC patients [76]. Peritumoral injections of IL-2 have been shown to increase T-cell cytotoxicity within the tumor microenvironment as well as increase CTL and NK cells in

3

the peripheral blood of unresectable HNSCC patients [77]. Consistent with this observation, a Phase 3 randomized study of oral cavity and oropharynx SCC patients treated with perilymphatic IL-2 and surgery followed by radiation had a greater than 25% improvement in overall survival at 5 years [78]. In contrast, a Phase II trial of systemic IL-2 in refractory nasopharyngeal carcinoma was ineffective [79]. The difference in outcomes between these two studies could potentially be related to the mode of delivery, with systemic delivery resulting in more toxicity and/or less efficacy than perilymphatic delivery of IL-2. Low-dose perilymphatic IL-2 has also been used with success in combination with other cytokines. IRX-2 is a cell-derived, cytokine mixture of physiologic levels of IL-1, IL-2, IL-6, IL-8, tumor necrosis factor-alpha, interferon-gamma, G-CSF, and GM-CSF that is produced by stimulating human PBMCs with phytohemagglutinin under GMP conditions [80]. In combination with the immuneadjuvants cyclophosphamide and indomethacin, a Phase II study of IRX-2 in 27 previously untreated, resectable patients with Stage II-IV HNSCC showed promising results when given preoperatively. Tumor responses were seen in 16% of patients, and 74% of patients had either decreased or stable tumor size prior to definitive surgery [81]. Importantly, significant increases in tumor and regional lymphatic lymphocytic infiltration were observed in the IRX-treated cohort. Improved survival at 5 years was correlated with high overall lymphocyte infiltration in the tumor specimens [82]. Currently, a large multi-site international Phase 3 study is underway to validate these findings. Other single agent cytokines have been used in HNSCC patients, but on a limited basis with little clinical trial data available. A small Phase1/2 study of interferon-gamma was undertaken in 8 patients. Immunologic responses were evident in almost 50% of the patients with clinically measurable responses in 3 patients and stable disease in 4 patients [83]. Interferon-alpha has also been used in several small series in combination with cisplastin/5-fluorouracil [84], isotretinoin/vitamin E [85], and IL-2 [86] with mixed results. Interleukin-12 was studied in 10 patients with untreated HNSCC [87]. NK and B cell infiltration was increased in tumors and higher NK levels (CD56+ cells) of HNSCC patients, irrespective of IL-12 treatment, had a better overall survival than those with a low number.

Therapeutic cancer vaccines The goal of cancer vaccines is to generate clinically meaningful tumor regression through the activation of the adaptive immune system with the presentation of tumor–associated/specific antigens (TAA/TSA) in the context of a peptide–major histocompatibility complex (MHC) for T-cell recognition or on the surface of the tumor cell for B-cell recognition [88]. TAAs are also present on normal tissues, in contrast to TSA which are unique to the tumor, and therefore tend to be weekly immunogenic. Cancer vaccines seek to break this tolerant phenotype through several different vaccine strategies that have been tried in Phase 2 and 3 clinical trials: presentation of peptides or proteins with adjuvant, recombinant viruses, bacteria or yeast, whole tumor cells, or delivery of proteinor peptide-activated dendritic cells (DCs) [88]. The majority of clinical studies have been performed in melanoma, glioma, as well as prostate, colorectal, and pancreatic carcinoma as well as in HNSCC. DC vaccines take advantage of the potent antigen presenting capacity of DCs to stimulate primary and secondary T-cell and Bcell immune responses. In HNSCC, targeting mutated proteins in the p53 pathway has been an area of active investigation given the prevalence of these mutations in this patient population. A Phase 1b clinical trial of 16 patients with mostly advanced HNSCC (14/16) were treated with 3 interval injections of dendritic cells pulsed with wildtype (wt) p53 peptides [89]. There was no

Please cite this article in press as: Li Q et al. Immunotherapy for head and neck squamous cell carcinoma. Oral Oncol (2015), http://dx.doi.org/10.1016/ j.oraloncology.2014.12.005

4

Q. Li et al. / Oral Oncology xxx (2015) xxx–xxx

observed Grade 2–4 toxicity and 2-year disease free survival (DFS) was 88%. Importantly, only 4/16 patients had increased IFNgamma when presented with p53 peptide in Elispot assays, but p53-specific T-cell frequencies were observed in 11/16 patients. Interestingly, immune-inhibitory regulatory T (Treg) cells were decreased in vaccinated patients. Human papilloma virus (HPV) has been another target of cancer vaccine strategies in HNSCC. HPV is thought to be important in the pathogenesis of oropharyngeal SCC and is found in the majority of tumors of this subtype [90]. The oncoproteins E6 and/or E7 have been targets in both cervical malignancies as well as HPV+ HNSCC. A feasibility study of a peptide vaccine against HPV16 peptide was performed in 5 patients using a Trojan vaccine [91]. Four patients had a measurable immune response to relevant HLA-II peptides but not to HLA-I restricted peptides. Larger studies targeting E6/ E7 have been performed in cervical [92] and vulvar [93] intraepithelial neoplasia with encouraging clinical results. Adoptive T cell immunotherapy of HNSCC Adoptive T cell immunotherapy is a technique that involves the administration of externally manipulated T cells that have been derived from tumor infiltrating lymphocytes (TIL), ex-vivo modified T cells with engineered T cell receptors, or chimeric antigen receptor that links the T cell response with an external antibody to relevant antigen [38]. This approach was studied in six patients with HNSCC at the University of Michigan. Activated vaccineprimed lymph node (VPLN) cells were administered in conjunction with IL-2. Anti-CD3-activated VPLN cells secreted IFN-gamma and GM-CSF in response to autologous tumor cells but no significant tumor responses were noted. One of the main limitations of this approach, along with other vaccine approaches, has been the low frequency of tumor-reactive T cells generated in treated patients. Rosenberg and his colleagues have potentially found a technique to bypass this problem [94]. Using whole-exomic sequencing, they identified a point mutation that can be recognized by a Th1 population within the TIL and this population is expanded ex-vivo and adoptively transferred back to the patient. Tumor regression was observed for over 6 months along with the identification of polyfunctional Th1 cells in the peripheral blood. Immunological targeting of HNSCC cancer stem cells One promising area is targeting tumor-initiating or cancer stem cells (CSCs). CSC population in HNSCC is linked to treatment failure, recurrence and metastasis [95]. Work originally performed by Clarke et al. revealed the existence of a small subpopulation of highly tumorigenic cells in breast cancer that were given the label cancer stem cells [96]. Following this report, our group demonstrated that the cell surface marker CD44 could be used to identify the highly tumorigenic population of cells from HNSCC [97]. Subsequently, we showed that high levels of aldehyde dehydrogenase (ALDHhigh) activity is an even more specific marker for the CSC population in HNSCC [98]. Importantly, in vitro CSC-primed T cells have been shown to specifically target CSCs in vivo. CSC-specific CD8+ T cells were generated from human acute myeloid leukemia (AML) stem cells in 1999 by Bonnet et al., and have been proven to mediate tumor regression after injection into NOD/SCID mice [99]. In addition, Brown and colleagues isolated CD133+ brain tumor stem cells (BTSCs), and demonstrated that these BTSCs are susceptible to perforin-dependent CTL-mediated cytolysis [100]. In a more relevant study, Visus and colleagues generated aldehyde dehydrogenase 1 family member A1 (ALDHA1)bright CSC-specific CD8+ T cells by using antigenic peptide from ALDH1A1. They demonstrated that transfer of ALDH1A1-specific CD8+ T cells

eliminated ALDHA1bright cancer stem cells from head and neck squamous cell carcinoma, inhibited tumor growth and metastases, and prolonged survival of xenograft-bearing immunodeficient mice [101,102]. Our group recently generated an anti-CD3/antiCD133 bispecific antibody (BsAb) and bound it to the cytokineinduced killer (CIK) cells as effector cells (BsAb-CIK) to target CD133high CSCs. The killing of CD133high cancer stem cells by the BsAb-CIK cells was significantly higher than the killing by the parental CIK or by CIK cells bound with anti-CD3 (CD3-CIK) without CD133 targeting both in vitro and in vivo [103]. Together, these studies suggest that potential CSC-specific T cells can be generated in vitro for subsequent adoptive transfer into tumor-bearing hosts to target CSCs in vivo and eradicate tumors including HNSCC. Specifically targeting cancer stem cells with a DC vaccine is also an innovative and rationale approach that could specifically address the most replicative and therapy-resistant subpopulation of cancer cells. Utilizing a murine squamous cell carcinoma tumor model (SCC7), we recently reported that the administration of dendritic cells pulsed with ALDHhigh tumor cells (the cancer stem cells) is significantly more effective as an antigen source in inducing protective anti-tumor immunity than whole tumor lysate or the nonCSC cell population [104]. We have also demonstrated similar findings in vitro when human HNSCC tumors are treated with autologous DC pulsed with cancer stem cells (manuscript under review). It is becoming increasingly clear that cancer vaccines for the majority of solid tumor malignancies, including HNSCC, will be the most beneficial in limited disease settings as either a neoadjuvant or adjuvant treatment rather than a stand alone therapy. Adding more T cells or APCs to an immunosuppressed systemic host or tumor microenvironment will unlikely show success as a single modality therapy. Combinatorial approaches will be needed along with cancer vaccines including reversing immunosuppression, chemoradiotherapy, or other synergistic therapies. Conclusion The role of the immune system in the initiation and progression of carcinogenesis in HNSCC is only beginning to be understood. Immunosuppression in HNSCC has been known for many years, but the exact pathways are only now being elucidated. Taking advantage of this new information will undoubtedly allow clinicians to better care for this population by exploiting these pathways. Future clinical trials in HNSCC will need to consider immunotherapeutic strategies in conjunction with either standard approaches and/or other innovative therapies to achieve the best outcomes for this difficult to treat population. Conflict of interest statement The authors have no conflicts of interest as it relates to the information and/or data in this review manuscript. References [1] Kuss I, Hathaway B, Ferris RL, Gooding W, Whiteside TL. Imbalance in absolute counts of T lymphocyte subsets in patients with head and neck cancer and its relation to disease. Adv Otorhinolaryngol 2005;62:161–72. [2] Schaefer C, Kim GG, Albers A, Hoermann K, Myers EN, Whiteside TL. Characteristics of CD4+CD25+ regulatory T cells in the peripheral circulation of patients with head and neck cancer. Br J Cancer 2005;92:913–20 [see comment]. [3] Strauss L, Bergmann C, Szczepanski M, Gooding W, Johnson JT, Whiteside TL. A unique subset of CD4+CD25highFoxp3+ T cells secreting interleukin-10 and transforming growth factor-beta1 mediates suppression in the tumor microenvironment. Clin Cancer Res 2007;13:4345–54. [4] Li C, Shintani S, Terakado N, Nakashiro K, Hamakawa H. Infiltration of tumorassociated macrophages in human oral squamous cell carcinoma. Oncol Rep 2002;9:1219–23.

Please cite this article in press as: Li Q et al. Immunotherapy for head and neck squamous cell carcinoma. Oral Oncol (2015), http://dx.doi.org/10.1016/ j.oraloncology.2014.12.005

Q. Li et al. / Oral Oncology xxx (2015) xxx–xxx [5] Grandis JR, Falkner DM, Melhem MF, Gooding WE, Drenning SD, Morel PA. Human leukocyte antigen class I allelic and haplotype loss in squamous cell carcinoma of the head and neck: clinical and immunogenetic consequences. Clin Cancer Res 2000;6:2794–802. [6] Whiteside TL. Immunobiology of head and neck cancer. Cancer Metastasis Rev 2005;24:95–105 [Review] [39 refs]. [7] Gabrilovich DI, Chen HL, Girgis KR. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med 1996 Nov;1996(2):1096–103 [erratum appears in Nat Med 1996 Nov;2(11):1267]. [8] Benefield J, Petruzzelli GJ, Fowler S, Taitz A, Kalkanis J, Young MR. Regulation of the steps of angiogenesis by human head and neck squamous cell carcinomas. Invasion Metastasis 1996;16:291–301. [9] Schroeder CP, Yang P, Newman RA, Lotan R. Eicosanoid metabolism in squamous cell carcinoma cell lines derived from primary and metastatic head and neck cancer and its modulation by celecoxib. Cancer Biol Ther 2004;3:847–52 [see comment]. [10] Qin H, Valentino J, Manna S, et al. Gene therapy for head and neck cancer using vaccinia virus expressing IL-2 in a murine model, with evidence of immune suppression. Mol Ther: J Am Soc Gene Ther 2001;4:551–8. [11] Dasgupta S, Bhattacharya-Chatterjee M, O’Malley BW, Chatterjee SK. Inhibition of NK cell activity through TGF-beta 1 by down-regulation of NKG2D in a murine model of head and neck cancer. J Immunol 2005;175:5541–50. [12] Bauernhofer T, Kuss I, Henderson B, Baum AS, Whiteside TL. Preferential apoptosis of CD56dim natural killer cell subset in patients with cancer. Eur J Immunol 2003;33:119–24. [13] Almand B, Resser JR, Lindman B, et al. Clinical significance of defective dendritic cell differentiation in cancer. Clin Cancer Res 2000;6:1755–66. [14] Whiteside TL, Stanson J, Shurin MR, Ferrone S. Antigen-processing machinery in human dendritic cells: up-regulation by maturation and down-regulation by tumor cells. J Immunol 2004;173:1526–34. [15] Ferris RL, Whiteside TL, Ferrone S. Immune escape associated with functional defects in antigen-processing machinery in head and neck cancer. Clin Cancer Res 2006;12:3890–5. [16] Meissner M, Reichert TE, Kunkel M, et al. Defects in the human leukocyte antigen class I antigen processing machinery in head and neck squamous cell carcinoma: association with clinical outcome. Clin Cancer Res 2005;11:2552–60. [17] Bandoh N, Ogino T, Katayama A, et al. HLA class I antigen and transporter associated with antigen processing downregulation in metastatic lesions of head and neck squamous cell carcinoma as a marker of poor prognosis. Oncol Rep 2010;23:933–9. [18] Mole RH. Whole body irradiation; radiobiology or medicine? Br J Radiol 1953;26:234–41. [19] Demaria S, Ng B, Devitt ML, et al. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys 2004;58:862–70. [20] Postow MA, Callahan MK, Barker CA, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med 2012;366:925–31. [21] Stamell EF, Wolchok JD, Gnjatic S, Lee NY, Brownell I. The abscopal effect associated with a systemic anti-melanoma immune response. Int J Radiat Oncol Biol Phys 2013;85:293–5. [22] Apetoh L, Ghiringhelli F, Tesniere A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med 2007;13:1050–9. [23] André N, Carré M, Pasquier E. Metronomics: towards personalized chemotherapy? Nat Rev Clin Oncol 2014;11:413–31. [24] Tesniere A, Apetoh L, Ghiringhelli F, et al. Immunogenic cancer cell death: a key-lock paradigm. Curr Opin Immunol 2008;20:504–11. [25] Kaneno R, Shurin GV, Tourkova IL, Shurin MR. Chemomodulation of human dendritic cell function by antineoplastic agents in low noncytotoxic concentrations. J Transl Med 2009;7:58. [26] Kaneno R, Shurin GV, Kaneno FM, Naiditch H, Luo J, Shurin MR. Chemotherapeutic agents in low noncytotoxic concentrations increase immunogenicity of human colon cancer cells. Cell Oncol (Dordr) 2011;34:97–106. [27] Ghiringhelli F, Menard C, Puig PE, et al. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol Immunother 2007;56:641–8. [28] Banissi C, Ghiringhelli F, Chen L, Carpentier AF. Treg depletion with a lowdose metronomic temozolomide regimen in a rat glioma model. Cancer Immunol Immunother 2009;58:1627–34. [29] Zhao J, Cao Y, Lei Z, Yang Z, Zhang B, Huang B. Selective depletion of CD4+CD25+Foxp3+ regulatory T cells by low-dose cyclophosphamide is explained by reduced intracellular ATP levels. Cancer Res 2010;70:4850–8. [30] Kan S, Hazama S, Maeda K, et al. Suppressive effects of cyclophosphamide and gemcitabine on regulatory T-cell induction in vitro. Anticancer Res 2012;32:5363–9. [31] Michels T, Shurin GV, Naiditch H, Sevko A, Umansky V, Shurin MR. Paclitaxel promotes differentiation of myeloid-derived suppressor cells into dendritic cells in vitro in a TLR4-independent manner. J Immunotoxicol 2012;9:292–300. [32] Geary SM, Lemke CD, Lubaroff DM, Salem AK. The combination of a low-dose chemotherapeutic agent, 5-fluorouracil, and an adenoviral tumor vaccine has

[33]

[34]

[35]

[36]

[37]

[38]

[39]

[40]

[41] [42]

[43]

[44]

[45]

[46] [47]

[48]

[49]

[50]

[51] [52]

[53]

[54]

[55]

[56]

[57]

[58]

5

a synergistic benefit on survival in a tumor model system. PLoS ONE 2013;8:e67904. Todaro M, Meraviglia S, Caccamo N, Stassi G, Dieli F. Combining conventional chemotherapy and cd T cell-based immunotherapy to target cancer-initiating cells. Oncoimmunology 2013;2:e25821. Moyer JS, Maine G, Mule JJ. Early vaccination with tumor-lysate-pulsed dendritic cells after allogeneic bone marrow transplantation has antitumor effects. Biol Blood Marrow Transpl 2006;12:1010–9. Moyer JS, Li J, Wei S, Teitz-Tennenbaum S, Chang AE. Intratumoral dendritic cells and chemoradiation for the treatment of murine squamous cell carcinoma. J Immunother 2008;31:885–95. Teitz-Tennenbaum S, Li Q, Rynkiewicz S, et al. Radiotherapy potentiates the therapeutic efficacy of intratumoral dendritic cell administration. Cancer Res 2003;63:8466–75. Teitz-Tennenbaum S, Li Q, Okuyama R, et al. Mechanisms involved in radiation enhancement of intratumoral dendritic cell therapy. J Immunother 2008;31:345–58. Raval RR, Sharabi AB, Walker AJ, Drake CG, Sharma P. Tumor immunology and cancer immunotherapy: summary of the 2013 SITC primer. J Immunother Cancer 2014;2:14. Turksma AW, Braakhuis BJ, Bloemena E, Meijer CJ, Leemans CR, Hooijberg E. Immunotherapy for head and neck cancer patients: shifting the balance. Immunotherapy 2013;5:49–61. Gildener-Leapman N, Ferris RL, Bauman JE. Promising systemic immunotherapies in head and neck squamous cell carcinoma. Oral Oncol 2013;49:1089–96. Grandis JR, Tweardy DJ. TGF-alpha and EGFR in head and neck cancer. J Cell Biochem Suppl 1993;17F:188–91. Licitra L, Mesia R, Rivera F, et al. Evaluation of EGFR gene copy number as a predictive biomarker for the efficacy of cetuximab in combination with chemotherapy in the first-line treatment of recurrent and/or metastatic squamous cell carcinoma of the head and neck: EXTREME study. Ann Oncol 2011;22:1078–87. Licitra L, Störkel S, Kerr KM, et al. Predictive value of epidermal growth factor receptor expression for first-line chemotherapy plus cetuximab in patients with head and neck and colorectal cancer: analysis of data from the EXTREME and CRYSTAL studies. Eur J Cancer 2013;49:1161–8. Martins RG, Parvathaneni U, Bauman JE, et al. Cisplatin and radiotherapy with or without erlotinib in locally advanced squamous cell carcinoma of the head and neck: a randomized phase II trial. J Clin Oncol 2013;31:1415–21. Argiris A, Ghebremichael M, Gilbert J, et al. Phase III randomized, placebocontrolled trial of docetaxel with or without gefitinib in recurrent or metastatic head and neck cancer: an eastern cooperative oncology group trial. J Clin Oncol 2013;31:1405–14. Bauman JE, Ferris RL. Integrating novel therapeutic monoclonal antibodies into the management of head and neck cancer. Cancer 2014;120:624–32. Bonner JA, Harari PM, Giralt J, et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med 2006;354:567–78. Basavaraj C, Sierra P, Shivu J, Melarkode R, Montero E, Nair P. Nimotuzumab with chemoradiation confers a survival advantage in treatment-naïve head and neck tumors over expressing EGFR. Cancer Biol Ther 2010;10:673–81. Reddy BK, Lokesh V, Vidyasagar MS, et al. Nimotuzumab provides survival benefit to patients with inoperable advanced squamous cell carcinoma of the head and neck: a randomized, open-label, phase IIb, 5-year study in Indian patients. Oral Oncol 2014;50:498–505. Vermorken JB, Stöhlmacher-Williams J, Davidenko I, et al. Cisplatin and fluorouracil with or without panitumumab in patients with recurrent or metastatic squamous-cell carcinoma of the head and neck (SPECTRUM): an open-label phase 3 randomised trial. Lancet Oncol 2013;14:697–710. Vermorken JB, Mesia R, Rivera F, et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl J Med 2008;359:1116–27. Srivastava RM, Lee SC, Andrade Filho PA, et al. Cetuximab-activated natural killer and dendritic cells collaborate to trigger tumor antigen-specific T-cell immunity in head and neck cancer patients. Clin Cancer Res 2013;19:1858–72. Teknos TN, Cox C, Yoo S, et al. Elevated serum vascular endothelial growth factor and decreased survival in advanced laryngeal carcinoma. Head Neck 2002;24:1004–11. Kyzas PA, Cunha IW, Ioannidis JP. Prognostic significance of vascular endothelial growth factor immunohistochemical expression in head and neck squamous cell carcinoma: a meta-analysis. Clin Cancer Res 2005;11:1434–40. Fury MG, Lee NY, Sherman E, et al. A phase 2 study of bevacizumab with cisplatin plus intensity-modulated radiation therapy for stage III/IVB head and neck squamous cell cancer. Cancer 2012;118:5008–14. Yao M, Galanopoulos N, Lavertu P, et al. A phase II study of bevacizumab in combination with docetaxel and radiation in locally advanced squamous cell carcinoma of the head and neck. Head Neck 2014. Argiris A, Kotsakis AP, Hoang T, et al. Cetuximab and bevacizumab: preclinical data and phase II trial in recurrent or metastatic squamous cell carcinoma of the head and neck. Ann Oncol 2013;24:220–5. Bellati F, Napoletano C, Ruscito I, et al. Complete remission of ovarian cancer induced intractable malignant ascites with intraperitoneal bevacizumab. Immunological observations and a literature review. Invest New Drugs 2010;28:887–94.

Please cite this article in press as: Li Q et al. Immunotherapy for head and neck squamous cell carcinoma. Oral Oncol (2015), http://dx.doi.org/10.1016/ j.oraloncology.2014.12.005

6

Q. Li et al. / Oral Oncology xxx (2015) xxx–xxx

[59] Mansfield AS, Nevala WK, Lieser EA, Leontovich AA, Markovic SN. The immunomodulatory effects of bevacizumab on systemic immunity in patients with metastatic melanoma. Oncoimmunology 2013;2:e24436. [60] Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363:711–23. [61] Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12:252–64. [62] Strauss L, Bergmann C, Gooding W, Johnson JT, Whiteside TL. The frequency and suppressor function of CD4+CD25highFoxp3+ T cells in the circulation of patients with squamous cell carcinoma of the head and neck. Clin Cancer Res 2007;13:6301–11. [63] Jie HB, Gildener-Leapman N, Li J, et al. Intratumoral regulatory T cells upregulate immunosuppressive molecules in head and neck cancer patients. Br J Cancer 2013;109:2629–35. [64] Bauman JE, Gooding WE, Clump DA, et al. Phase I trial of cetuximab, intensity modulated radiotherapy (IMRT), and the anti-CTLA-4 monoclonal antibody (mAb) ipilimumab in previously untreated, locally advanced head and neck squamous cell carcinoma (PULA HNSCC). J Clin Oncol 2014:32 [suppl; abstr TPS6104]. [65] Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol 2008;8:467–77. [66] Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7–H1 promotes Tcell apoptosis: a potential mechanism of immune evasion. Nat Med 2002;8:793–800. [67] Cho YA, Yoon HJ, Lee JI, Hong SP, Hong SD. Relationship between the expressions of PD-L1 and tumor-infiltrating lymphocytes in oral squamous cell carcinoma. Oral Oncol 2011;47:1148–53. [68] Lyford-Pike S, Peng S, Young GD, et al. Evidence for a role of the PD-1:PD-L1 pathway in immune resistance of HPV-associated head and neck squamous cell carcinoma. Cancer Res 2013;73:1733–41. [69] Malm IJ, Bruno TC, Fu J, et al. Expression profile and in vitro blockade of PD-1 in HPV-negative head and neck squamous cell carcinoma. Head Neck 2014. [70] Curiel TJ, Wei S, Dong H, et al. Blockade of B7–H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med 2003;9:562–7. [71] Kuang DM, Zhao Q, Peng C, et al. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med 2009;206:1327–37. [72] Liu Y, Zeng B, Zhang Z, Zhang Y, Yang R. B7–H1 on myeloid-derived suppressor cells in immune suppression by a mouse model of ovarian cancer. Clin Immunol 2008;129:471–81. [73] Brahmer JR, Drake CG, Wollner I, et al. Phase I study of single-agent antiprogrammed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 2010;28:3167–75. [74] Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 2014. [75] Seiwert TY, Burtness B, Weiss J, et al. A phase Ib study of MK-3475 in patients with human papillomavirus (HPV)-associated and non-HPV–associated head and neck (H/N) cancer. J Clin Oncol 2014;32 [suppl; abstr 6011]. [76] Grande C, Firvida JL, Navas V, Casal J. Interleukin-2 for the treatment of solid tumors other than melanoma and renal cell carcinoma. Anticancer Drugs 2006;17:1–12. [77] Whiteside TL, Letessier E, Hirabayashi H, et al. Evidence for local and systemic activation of immune cells by peritumoral injections of interleukin 2 in patients with advanced squamous cell carcinoma of the head and neck. Cancer Res 1993;53:5654–62. [78] De Stefani A, Forni G, Ragona R, et al. Improved survival with perilymphatic interleukin 2 in patients with resectable squamous cell carcinoma of the oral cavity and oropharynx. Cancer 2002;95:90–7. [79] Chi KH, Myers JN, Chow KC, et al. Phase II trial of systemic recombinant interleukin-2 in the treatment of refractory nasopharyngeal carcinoma. Oncology 2001;60:110–5. [80] Egan JE, Quadrini KJ, Santiago-Schwarz F, Hadden JW, Brandwein HJ, Signorelli KL. IRX-2, a novel in vivo immunotherapeutic, induces maturation and activation of human dendritic cells in vitro. J Immunother 2007;30:624–33. [81] Wolf GT, Fee WE, Dolan RW, et al. Novel neoadjuvant immunotherapy regimen safety and survival in head and neck squamous cell cancer. Head Neck 2011;33:1666–74. [82] Berinstein NL, Wolf GT, Naylor PH, et al. Increased lymphocyte infiltration in patients with head and neck cancer treated with the IRX-2 immunotherapy regimen. Cancer Immunol Immunother 2012;61:771–82.

[83] Richtsmeier WJ, Koch WM, McGuire WP, Poole ME, Chang EH. Phase I-II study of advanced head and neck squamous cell carcinoma patients treated with recombinant human interferon gamma. Arch Otolaryngol Head Neck Surg 1990;116:1271–7. [84] Bazarbashi S, Rahal M, Raja MA, et al. A pilot trial of combination cisplatin, 5fluorouracil and interferon-alpha in the treatment of advanced esophageal carcinoma. Chemotherapy 2002;48:211–6. [85] Seixas-Silva JA, Richards T, Khuri FR, et al. Phase 2 bioadjuvant study of interferon alfa-2a, isotretinoin, and vitamin E in locally advanced squamous cell carcinoma of the head and neck: long-term follow-up. Arch Otolaryngol Head Neck Surg 2005;131:304–7. [86] Urba SG, Forastiere AA, Wolf GT, Amrein PC. Intensive recombinant interleukin-2 and alpha-interferon therapy in patients with advanced head and neck squamous carcinoma. Cancer 1993;71:2326–31. [87] van Herpen CM, van der Laak JA, de Vries IJ, et al. Intratumoral recombinant human interleukin-12 administration in head and neck squamous cell carcinoma patients modifies locoregional lymph node architecture and induces natural killer cell infiltration in the primary tumor. Clin Cancer Res 2005;11:1899–909. [88] Schlom J, Hodge JW, Palena C, et al. Therapeutic cancer vaccines. Adv Cancer Res 2014;121:67–124. [89] Schuler PJ, Harasymczuk M, Visus C, et al. Phase I dendritic cell p53 peptide vaccine for head and neck cancer. Clin Cancer Res 2014;20:2433–44. [90] D’Souza G, Kreimer AR, Viscidi R, et al. Case-control study of human papillomavirus and oropharyngeal cancer. N Engl J Med 2007;356:1944–56. [91] Voskens CJ, Sewell D, Hertzano R, et al. Induction of MAGE-A3 and HPV-16 immunity by Trojan vaccines in patients with head and neck carcinoma. Head Neck 2012;34:1734–46. [92] Brun JL, Dalstein V, Leveque J, et al. Regression of high-grade cervical intraepithelial neoplasia with TG4001 targeted immunotherapy. Am J Obstet Gynecol 2011;204. 169.e1–8. [93] Kenter GG, Welters MJ, Valentijn AR, et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med 2009;361:1838–47. [94] Tran E, Turcotte S, Gros A, et al. Cancer immunotherapy based on mutationspecific CD4+ T cells in a patient with epithelial cancer. Science 2014;344:641–5. [95] Davis SJ, Divi V, Owen JH, et al. Metastatic potential of cancer stem cells in head and neck squamous cell carcinoma. Arch Otolaryngol Head Neck Surg 2010;136:1260–6. [96] Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA 2003;100:3983–8. [97] Prince ME, Sivanandan R, Kaczorowski A, et al. Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma. Proc Natl Acad Sci USA 2007;104:973–8. [98] Clay MR, Tabor M, Owen JH, et al. Single-marker identification of head and neck squamous cell carcinoma cancer stem cells with aldehyde dehydrogenase. Head Neck 2010;32:1195–201. [99] Bonnet D, Warren EH, Greenberg PD, Dick JE, Riddell SR. CD8(+) minor histocompatibility antigen-specific cytotoxic T lymphocyte clones eliminate human acute myeloid leukemia stem cells. Proc Natl Acad Sci USA 1999;96:8639–44. [100] Brown CE, Starr R, Martinez C, et al. Recognition and killing of brain tumor stem-like initiating cells by CD8+ cytolytic T cells. Cancer Res 2009;69:8886–93. [101] Visus C, Wang Y, Lozano-Leon A, et al. Targeting ALDH(bright) human carcinoma-initiating cells with ALDH1A1-specific CD8+ T cells. Clin Cancer Res 2011;17:6174–84. [102] Visus C, Ito D, Amoscato A, et al. Identification of human aldehyde dehydrogenase 1 family member A1 as a novel CD8+ T-cell-defined tumor antigen in squamous cell carcinoma of the head and neck. Cancer Res 2007;67:10538–45. [103] Huang J, Li C, Wang Y, et al. Cytokine-induced killer (CIK) cells bound with anti-CD3/anti-CD133 bispecific antibodies target CD133(high) cancer stem cells in vitro and in vivo. Clin Immunol 2013;149:156–68. [104] Ning N, Pan Q, Zheng F, et al. Cancer stem cell vaccination confers significant antitumor immunity. Cancer Res 2012;72:1853–64.

Please cite this article in press as: Li Q et al. Immunotherapy for head and neck squamous cell carcinoma. Oral Oncol (2015), http://dx.doi.org/10.1016/ j.oraloncology.2014.12.005

Immunotherapy for head and neck squamous cell carcinoma.

To review the current state of immunotherapy of head and neck squamous cell carcinoma...
407KB Sizes 0 Downloads 14 Views