Accepted Manuscript Title: Overcoming immunosuppression in bone metastases Authors: Zachary Reinstein, Sahithi Pamarthy, Vinay Sagar, Ricardo Costa, Sarki A. Abdulkadir, Francis J. Giles, Benedito A. Carneiro PII: DOI: Reference:
S1040-8428(17)30179-8 http://dx.doi.org/doi:10.1016/j.critrevonc.2017.05.004 ONCH 2385
To appear in:
Critical Reviews in Oncology/Hematology
Received date: Revised date: Accepted date:
13-3-2017 30-4-2017 9-5-2017
Please cite this article as: Reinstein Zachary, Pamarthy Sahithi, Sagar Vinay, Costa Ricardo, Abdulkadir Sarki A, Giles Francis J, Carneiro Benedito A.Overcoming immunosuppression in bone metastases.Critical Reviews in Oncology and Hematology http://dx.doi.org/10.1016/j.critrevonc.2017.05.004 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Overcoming immunosuppression in bone metastases Zachary Reinstein1,2,3, Sahithi Pamarthy1,2, Vinay Sagar1,2, Ricardo Costa 1, Sarki A Abdulkadir2,4,5, Francis J Giles 1, Benedito A Carneiro1* 1
Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, USA. 2 The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA 3 Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah 86402, USA 4 Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA 5 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
*
Corresponding author: Benedito A Carneiro, MD, MS Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University 233 E Superior St Olson Pavilion Chicago, IL 60611 Phone: 312-472-1234 Fax: 312-472-0564 E-mail:
[email protected]
Keywords: Bone metastases; immunosuppression; bone metastatic disease; skeletal related events Running title: Immunosuppression in bone metastases Contributions: ZR wrote, reviewed and revised the manuscript and tables, SP wrote, reviewed, and revised the manuscript and figure, VS contributed to the figure and reviewed/revised the manuscript. RC wrote, reviewed, and revised the manuscript. SAA, FJG, BAC conceived, reviewed and revised the manuscript. Acknowledgements: We would like to thank The Woman’s Board of Northwestern Memorial Hospital for their continued support.
1
ABSTRACT
Bone metastases are present in up to 70% of advanced prostate and breast cancers and occur at significant rates in a variety of other cancers. Bone metastases can be associated with significant morbidity. The establishment of bone metastasis activates several immunosuppressive mechanisms. Hence, understanding the tumor-bone microenvironment is crucial to inform the development of novel therapies. This review describes the current standard of care for patients with bone metastatic disease and novel treatment options targeting the microenvironment. Treatments reviewed include immunotherapies, cryoablation, and targeted therapies. Combinatorial treatment strategies including targeted therapies and immunotherapies shows promise in pre-clinical and clinical studies to overcome the suppressive environment and improve treatment of bone metastases.
1. Introduction
Prevention and effective treatment of bone metastatic disease can have a significant impact in the outcomes of patients with advanced malignancies. The burden of bone metastatic disease is demonstrated by autopsy studies identifying bone metastases in approximately 70 % of breast and prostate cancer, 30-40 % of non-small cell lung cancer (NSCLC), and 20-35 % of thyroid and kidney cancer-related deaths [1]. It has been estimated that greater than 50 % of patients with bone metastases die within 3 years of diagnosis [2]. Furthermore, bone metastases are associated with skeletal related events (SRE) including pathological fractures, spinal cord compression, and hypercalcemia, which severely compromise patient quality of life (QoL) [3,4]. Even though most patients have concomitant metastases in visceral organs and lymph nodes, there is a subset of patients with bone-predominant metastatic disease that may require a distinct treatment approach compared to, for instance, patients with only pulmonary metastases based on site-specific impact on disease prognosis and, possibly, distinct patterns of response to systemic therapies such as chemotherapy and immunotherapies. Therefore, advancing the understanding of mechanisms involved in the establishment of bone metastases
2
can lead to the identification of therapeutic targets with meaningful implication in the management of these patients. This manuscript reviews the challenges in treating bone metastases and highlights emerging therapeutic strategies exploring the interface between the bone microenvironment and immune system with the ultimate goal of fostering the advancement of novel bone-directed therapies.
2. Bone tumor microenvironment 2.1 Bone metastatic niche Tumor cells ‘seed’ the bone marrow in a multistep process [5]. Initially, circulating tumor cells (CTCs) create a pre-metastatic niche with the production of factors that render the bone microenvironment conducive to tumor dissemination [6]. Disseminated tumor cells (DTCs) compete with “hematopoietic stem cell niche” in the bone marrow, thereby creating an “onconiche” where they can proliferate or remain dormant. The bone marrow niche seems to protect DTCs from immune surveillance contributing to resistance to chemotherapy and post treatment relapses by several mechanisms.
Bone marrow DTCs from breast cancer lose expression of Ki67 and overexpress ERBB2 that might play a role in resistance to chemotherapy. DTCs at distant sites found after chemotherapy also express cytokeratin heterodimers CK8/CK18 and CK8/CK19 known to inhibit major histocompatibility complex 1 (MHC 1) interactions with CD8+ T cells, a crucial step in immune escape [7]. To overcome natural killer (NK) cell cytotoxicity following loss of MHC, DTCs adopt a stem cell phenotype with reduced expression of NK cell receptor D (NKG2D) and MHC1 polypeptide related sequence (MICA/MICB) [8]. Furthermore, DTCs home to the hematopoietic stem cell (HSC) niche, which is the most hypoxic area of bone marrow. It is known that increased expression of hypoxia induced factor (HIF1 in neoplastic cells enhances a disintegrin and metalloproteinase (ADAM10) which in turn cleaves MICA/MICB leading to suppression of antigen presentation and NK cell cytotoxicity [9]. Therefore, the hypoxic environment of HSC niche in bone marrow contributes to immune escape, stem-like properties of DTCs, and promotes resistance to chemotherapy [10].
3
2.2 Osteomimicry It is well established that cancers like breast and prostate exhibit bone tropism and express bone associated genes and proteins, a phenomenon called “osteomimicry", which promotes their dissemination, survival, and proliferation in the bone [11]. Tumor cells homing to bone express molecules like osteonectin, cathepsin-K and connexins that are usually expressed by osteoclasts and osteoblasts. Prostate cancer cells acquire an osteoblast-like phenotype by expressing bone sialoprotein and osteocalccin. RUNX2 is a transcription factor crucial for osteoblast differentation and the expression of RUNX2 by cancer cells marks a key step in osteomimicry. The stem cell pathways Notch and Wnt were shown to be important for activation of hepatocyte growth factor (HGF) and its interaction with Met, another key pathway in osteomimicry [12]. Breast cancer cells also express osteoclast activating factors like parathyroid hormone-related protein (PTHrP) and TNF- to promote RANKL induced osteoclastogenesis [13]. Together, the expression of these molecules promotes colonization to bone, confers a survival advantage and initiates the tumor-bone vicious cycle.
2.3 Tumor-bone vicious cycle Bone is a significant site of metastatic disease because of its large surface area and high vascular supply. Tumor invasion to bone results in bone resorption and formation, a process induced by cancer cells and mediated by osteoblasts and osteoclasts [14]. Certain types of solid tumors metastasize to bone and induce osteolytic (bone destructive) or osteoblastic (bone formative) phenotypes. For example, metastases from prostate cancer frequently form osteoblastic lesions in contrast to the predominant osteolytic lesions associated with breast, lung and kidney cancers [15]. Tumor-derived signaling mediators like WNT, bone morphogenic proteins (BMPs) and transforming growth factor-beta (TGF-) stimulate osteoblast function by activating RUNX and activated transcription factor (ATF) signaling [16-18]. Activated osteoblasts secrete receptor activator of nuclear factor kappa (RANKL), which binds to its receptor RANK on osteoclast precursors and induces osteoclastogenesis through NF-kB, NFATc1 and C-JUN
4
signaling [19]. During the process of resorption, bone cells and the mineralized bone matrix release TGF-, IGF, and Ca2+ promoting tumor growth and release of osteolytic and osteoblastic factors like PTHrP, IL-6 and matrix metalloproteinases (MMPs) [20]. Thus, a “vicious cycle” of tumor-induced bone disease is formed, wherein tumor modifies the bone microenvironment to support its own survival [21].
2.4 Bone microenvironment – cellular and molecular mediators The vast immune cell composition of bone marrow including lymphocytes and myeloid cells exhibit both anti-tumor and tumor-promoting effects on bone metastases [22,23]. During an antitumor response, dendritic cells present tumor specific antigens to activate CD4+ T cells, which in turn activate CD8+ T cells leading to the killing of antigen positive tumor cells [24]. In contrast, activated regulatory T cells (T-Regs) and T helper cells type 17 (Th 17 cells) cause a protumor response through immune suppression and RANKL mediated osteoclast differentiation [25,26]. Additionally, myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) suppress T-cell mediated anti-tumor responses through production of cytokines and angiogenic factors [27,28]. In addition to the immune cells, endothelial cells, fibroblasts, and mesenchymal stem cells are recruited to promote neoangiogenesis and bone metastatic growth [29]. Tumor-derived exosomes were also shown to educate bone marrow progenitors to support tumor growth [30]. Together, the resident and infiltrated stromal cells and their molecular mediators constitute a unique bone metastatic microenvironment that results in sustained immunosuppression contributing to bone induced tumor growth (Figure 1). 2.5 Immune Evasion – mechanisms and implications Cancer cells thrive in the bone owing to their ability to escape immune surveillance by multiple mechanisms [31]. However, the role of the bone immune microenvironment in controlling or promoting metastatic progression remains open to further investigation. During normal physiological conditions, the combined CD4+ and CD8+ T cells constitute < 5% of mononuclear cells and NK cells make up to 1 -2% of lymphocyte populations. It is thought that metastatic
5
dissemination selects tumor cells that are resistant to apoptosis and hence are immune to T and NK cell mediated cytotoxicity [32]. Metastatic tumor cells routinely lose the expression of MHC class 1 antigen presentation leading to immune evasion. A majority of metastatic tumor cells target several molecules key to the immune checkpoint pathway to achieve immune suppression. Programmed cell death ligand (PD-L1) is expressed primarily on cancer cells and binds to the programmed cell death (PD)-1 receptor on T cells to inhibit their activation [33]. The recruitment of T-regs that express cytotoxic T lymphocyte associated antigen (CTLA 4), and lymphocyte activation gene-3 (LAG-3) leads to suppression of T cell activity [34]. Cytotoxic T cells and T regs also co-express T cell immunoglobulin and mucin containing domain (TIM 3) with PD-1, key surface membrane immune checkpoint proteins mediating immune suppression [35]. Hypoxic tumor cells secrete high amounts of adenosine, which inhibits T cell function by binding to A2AR, another immune checkpoint protein expressed on T cells [36,37].
Recent reports point to the loss of interferon regulatory factor 7 (IRF7), a key mediator of type 1 interferon gene signature, increases the risk of bone metastases [38]. Latent metastatic cells silence Wnt Signaling pathway to enter the quiescent state and escape innate immune responses for extended periods [39]. Similarly, suppression of JAK-STAT pathway impairs NK cell-mediated antitumor activity [40]. Tumor-derived growth factors and cytokines like TGF- and IL-6 recruit cancer-associated fibroblasts (CAFs), which target T-cell activity and also recruit tumor-associated macrophages to promote immune evasion. Activated MDSCs also mediate immune suppression through the production of reactive oxygen species that inhibits T-cell effector functions [27]. In addition to immune checkpoint inhibition, circulating tumor cells aggregate with platelets to escape immune recognition and TNF- related cell death. Supportively, platelets release pro-angiogenic factors that promote tumor metastasis [41]. Although the crosstalk between bone, tumor, and immune cells is well-known, bone metastasis frequently results in resistance to chemotherapy and post-treatment relapses due to immune escape by disseminated tumor cells. Detailed investigations in the field of tumor osteoimmunology are warranted to understand the mechanisms of immunosurveillance in tumor-induced bone disease [42] .
6
3. Standard of care treatment of bone metastases
Bone metastases are treated with the same approaches as metastatic disease involving other sites (i.e., chemotherapy, radiation therapy, and immunotherapy) with few exceptions of bonedirected therapy such as Radium-223 approved for metastatic prostate cancer. Osteoclast inhibitors such as bisphosphonates and denosumab are considered supportive therapies used to reduce the risk of SREs. Bisphosphates (i.e., zoledronic acid) stimulate osteoblasts and inhibit osteoclast differentiation and survival by binding to hydroxyapatite in the bone with selective inhibition of farnesyl pyrophosphate synthase in active areas of bone remodeling [43]. Denosumab, a fully human monoclonal antibody, binds to RANKL expressed on the surface of osteoblasts blocking the activation of the receptor (RANK) present on osteoclasts[44]. Denosumab disrupts the homeostasis of osteoblast and osteoclast activation and differentiation leading to a reduction of osteoclasts and osteoclast activity inhibition [45].
Zoledronic acid is usually given on every 4-week schedule however preliminary results from the CALGB 70604 phase 3 non-inferiority trial supported equivalent efficacy of two different dose schedules for the prevention of SRE among patients with metastatic cancer to the bones (i.e., breast cancer, prostate cancer, multiple myeloma) [46]. In light of the results of this trial zoledronic acid may be administered either monthly or every 3 months for patient with metastatic bone disease [47]. Approved by the FDA in 2013, denosumab was shown to significantly decrease skeletal-related events (SRE) compared to zoledronic acid. In addition, denosumab demonstrated fewer adverse events compared to zoledronic acid [48]. More recently, 3420 women with hormone-receptor positive breast cancer receiving adjuvant treatment with aromatase inhibitors were randomized to adjuvant treatment with denosumab vs. placebo every 6 months in the adjuvant denosumab in breast cancer trial (ABCSG-18). The patients who received denosumab had a significantly delayed time to first clinical fracture (HR 0.50, P<0.0001) [49]. After a median follow-up of 4 years, 203 disease free survival (DFS) events were observed in the placebo group, and 167 in the denosumab group (HR 0.81, P=0.051) [50].
7
Similar findings have been reported with adjuvant treatment with bisphosphonates [51]. These results suggest that bone-modifying agents not only reduce the incidence of SRE in the adjuvant treatment of breast cancer but may also have anti-tumor activity in a subset of patients with localized breast cancer.
Radiopharmaceuticals have also been used as a targeted therapy to bone metastases. These drugs act by releasing alpha, beta or gamma radiation to target tumor cells as the compounds accumulate in the bone matrix [52]. Both SR-89 and SM- 153 have been approved since the 1990’s and have shown to improve pain from bone metastases and reduce SREs [52]. Radium223 has been approved by the FDA for treatment of symptomatic metastatic CRPC with disease limited to the bones based on improvement in SREs, and pain when compared to SR-89/SM-153 but also was the first radiopharmaceutical to show an increase in overall survival [53]. Combination clinical studies of anti-androgens abiraterone and enzalutamide with Radium-223 and denosumab are ongoing [52].
In cancers with a high incidence of bone metastases, there are 3 classes of immunotherapies that have been approved. The first, immune checkpoint inhibitors include nivolumab (anti-PD1), ipilimumab (anti-CTLA-4), pembrolizumab (anti-PD-1), and atezolizumab (anti-PD-L1). All of these therapies are humanized antibodies against the immune checkpoints [54]. The next class is cytokine therapy which includes interferon , and interleukin-2. Both of these cytokines induce T-cell proliferation [55]. The dendritic cell therapy sipuleucel-T promotes stimulation of prostate cancer antigen specific T-cells [4]. While checkpoint inhibitors and other classes of immunotherapeutics have shown significant efficacy in controlling visceral metastatic disease in several malignancies, their efficacy specifically in bone metastases is not well understood. The bone-metastatic microenvironment displays a unique immune phenotype that could result in a distinct pattern of response to immune therapy when compared to other metastatic sites.
4. Emerging Therapies for Bone Metastases
8
4.1 Targeting the “Vicious Cycle”
Advances in targeted therapies have led to the approval of multiple small molecule inhibitors that target both tumor cells and the microenvironment. For the purposes of this review, the focus will be those targeted therapies, approved and in development, that specifically affect the tumor-bone microenvironment and immune suppression.
The "vicious cycle" of bone metastases involves the feedback loop between the bone-derived growth factors (RANKL, PTHrP, IL-1, IL-6, and IL-8) and tumor-derived osteolytic factors (TGF-, FGF, PDGF) [56]. Multiple therapies are being developed to target this cycle.
Osteoprotegerin (OPG) is a natural inhibitor of RANKL, and consequently, inhibits osteoclastogenesis. Yet, multiple experiments studying OPG administered systemically versus OPG produced by tumor cells at various concentrations showed conflicting results in regards to tumor progression and bone resorption [57-62]. Ryser et al devised a mathematical model of the OPG absorption gradient alongside a model of bone remodeling to determine the effects of OPG at different concentrations and demonstrated the optimal concentrations of OPG for therapeutic effect [63]. Additional research is necessary for validation and development of therapies.
Multiple strategies have been developed to inhibit TGF-with the most recent successes seen in small molecule inhibitors (galunisertib) and vaccinations (belagenpumatucel-L and gemogenovatucel-T) [64]. OS benefit was seen in phase II trials of galunisertib and belagenpumatucel-L. In addition, both of these treatments reduced TGF-and overall metastatic burden [65,66]. While bone metastases were not specifically measured in these studies, due to the crucial role of TGF- in the vicious cycle, these results should merit research in patients with bone metastases.
9
51 is an integrin that binds to fibronectin in the ECM and regulates tumor invasion and angiogenesis as mediated by (MMP-1) [67]. Thus 51 inhibition of the receptor in fibronectin prevents metastases and angiogenesis, especially in regards to the vicious cycle of osteolytic lesions [68]. Ac-PHSCN-NH2, a small peptide inhibitor showed no objective responses but showed progression-free survival in breast cancer patients for up to 14 months [69]. A second generation Ac-PhScN-NH2 inhibitor containing D-isomers of histidine (h) and cysteine (c)—is over 100,000-fold more potent at blocking basement membrane invasion by two breast cancer cells lines. Monotherapy in a mouse model reduced intratibial colony progression by almost 80 %[68].
P62 is a multi-functional protein that plays a significant anti-inflammatory role as well as induction of RANKL-stimulated osteoclastogenesis [70,71]. P62 is overexpressed in tumor tissues but not in normal tissue, making it an attractive target [72]. Because of this and the dual roles of P62, inhibition has been seen as a promising therapy to increase immune function in the tumor microenvironment and treatment of osteolytic lesions [72]. Three studies in animal models looking at the effectiveness of a DNA vaccine targeted against P62 saw preliminary antitumor activity with responses in tumor size and suppression of metastases. In addition, increased lymphocyte infiltration and suppression of osteoporosis was observed [71,73,74]. It has been hypothesized that in osteolytic tumors, these dual effects may prove this P62 vaccine highly effective [72].
Cabozantinib is a multi-targeted tyrosine kinase inhibitor which inhibits MET, vascular endothelial growth factor receptor 2 (VEGFR-2) as well as other tyrosine kinases including RET, KIT, AXL and FLT3, approved for medullary thyroid cancer and renal cell carcinoma [75]. MET is particularly overexpressed in bone metastases [76]. It has also been investigated in mCRPC after it showed significant inhibition of osteoblastic and osteolytic lesions in a mouse xenograft model [76]. Results in two, phase II trials also showed promising results, displaying an increase of progression-free survival from 5.9 weeks to 23.9 weeks compared to placebo, 63% bone scan response and pain palliation in 57% of patients [77,78]. These promising results led to two
10
large-scale phase III trials known at COMET I/II. Although an improvement in patient free survival (PFS) was noted, unfortunately, no overall survival advantage was observed in the larger cohort [79,80]. However, in the METEOR trial investigating cabozantinib as a second-line therapy in advanced renal cell carcinoma, OS survival benefit was observed when compared to everolimus [81]. Most interesting was that in patients with bone metastases, the hazard ratio was significantly reduced when compared to patients with no metastases. Choueiri et al hypothesize that this effect is due to cabozantinib specific action against bone metastases based on its ability to inhibit Met signaling, but further research is warranted before cabozantinib used as a specific bone-targeted therapy [81,82].
4.2 Targeting immunosupressive cells
As previously noted, TAMs play a crucial immunosuppressive role in the tumor microenvironment. Multiple drugs are being used or are in development to target TAMs. Interestingly, bisphosphates, like zoledronic acid, which inhibit osteoclasts to prevent bone reabsorption in osteolytic lesions, also play a similar role on TAMs [56]. Macrophages are of the same lineage as osteoclasts, and multiple studies point to zoledronic acid causing a change of TAM phenotype from the pro-tumoral M2 to the anti-tumoral M1 [56]. PLX3397 is another promising multi-targeted tyrosine kinase inhibitor in development that can target TAMs. PLX3397 inhibits colony stimulating factor 1 (CSF-1) which is responsible for recruiting immunosuppressive cells including M2 macrophages and MDSCs to the bone microenvironment [83]. Current preclinical data shows its efficacy in inhibiting the recruitment of immunosuppressive myeloid cells as well as improving the immune response in combination with other therapies [83-85]. Similarly, Bindarit targets chemokine (C-C motif) ligand 2 (CCL2), another cytokine responsible for macrophage recruitment. Preclinical studied have shown responses in animal models as well as the prevention of the establishment of the tumorstromal niche inherent to bone metastases [86,87].
11
Tasquinimod is second generation, quinoline-3-carboxamide, oral small molecule inhibitor that blocks the interactions of S100A9, a calcium and zinc binding protein involved in many inflammatory response pathways. It targets the tumor microenvironment by inhibition of angiogenesis and immune suppression mediated by TAMs, and prevention of the establishment of the bone-metastatic niche [88,89]. A phase II, randomized, double-blind trial of tasquinimod in mCRPC documented improvement of progression-free survival (PFS) (7.6 vs. 3.3 months, P=.0042) especially among patients with bone metastases (PFS 8.8 months) [90]. These results supported a phase III trial randomizing 1,245 patients with mCRPC to receive tasquinimod or placebo that showed a significant improvement of rPFS (7.0 vs. 4.4 months, HR= 0.64, P=0.001). No overall survival benefit was observed which decreased enthusiasm for further research with this drug [91].
Fibroblast are the key component of the immunosuppressive tumor stroma, therefore inhibitors targeting fibroblasts have significant therapeutic potential. Fibroblasts in idiopathic pulmonary fibrosis highly resemble cancer-associated fibroblasts (CAFs) with their expression of FAP. Thus, two recently approved drugs used to treat idiopathic pulmonary fibrosis, pirfenidone, and nintedanib, are being used to target CAFs in the tumor microenvironment, but results remain unpublished [36].
Although no current drugs have been developed to exclusively target immunosuppressive cells in the bone-tumor stroma, many current targeted therapies have off-target effects against immunosuppressive MDSCs and Tregs. TKIs like suntinib and imatinib block the STAT3 and IDO pathways. By inhibiting these pathways, maturation of MDSCs or Tregs is also inhibited thereby reducing their numbers in the bone-tumor microenvironment [92-95]. Bevacizumab, a VEGF antibody, plays a similar role by shifting monocyte development towards DCs instead of MDSCs in the bone marrow [96]. mTOR inhibitors like rapamycin and temsirolimus also act on the IDO pathway which in turn impairs the homeostasis and subsequent effectiveness of Tregs [97-99]. In addition, many of these target therapies have shown immunostimulating properties by acting on DCs and cytotoxic T-cells [100]. All of these techniques demonstrate a new treatment
12
paradigm of not just targeting the tumor cells, but also targeting the tumor-supportive microenvironment. Clinical studies have reflected this paradigm shift, with many studies exploring combinatorial approaches.
4.3 Increasing the Immune Response in Bone Metastases
In contrast to small molecule inhibitors, modulation of the immune system is also being investigated as a viable treatment option of bone metastases. Due to the unique immunoenvironment in bone metastases, adoptive cell transfer (ACT), vaccines, chimeric antigen receptor (CAR) therapies and other immunotherapies have been modified in order to increase efficacy in the bone metastatic microenvironment with promising results in various cancer models.
Methods to increase T-cell invasion into solid tumors have been developed and implemented for several years. Currently, adoptive cell transfer is only approved for melanoma and leukemia, but many other techniques are being developed to increased T-cell infiltration and efficacy with or without ex vivo expansion for bone metastases.
4.3.1 Adoptive cell transfer
Adoptive cell transfer (ACT) has been defined as “the administration of tumor-specific lymphocytes (obtained from the patient (autologous) or from a donor (allogeneic)) following a lymphodepleting preparative regimen” [101]. It holds promise for treatment of bone metastases, yet studies have shown that even after ACT T-cells in metastases still face heavy immunosuppression [102]. The most important method to improve the efficacy of ACT is lymphodepletion through whole-body irradiation or cytotoxic chemotherapy. Two studies of ACT in solid tumors saw complete response only after lymphodepletion protocols [103,104].
4.3.2 Dendritic Cell Therapy (Vaccines)
13
Dendritic cells are the primary promoter of a tumor-specific T-cell response through antigen presentation. Thus, several methods have been developed to improve on the immunogenicity of DCs in order to improve the anti-tumoral T-cell response and overcome bone metastatic immunosuppression.
With the recent approval of sipuleucel-T in prostate cancer, DC-based vaccines have been extensively researched to treat solid tumors and bone metastases. Vaccines can either be derived from (1) antigenic peptides of a tumor-associated antigen (TAA), like sipuleucel-T (2) whole tumor cells or (3) mRNA of TAAs [105]. In comparison with vaccines derived from antigenic peptides, those derived from whole tumor cells (either lysates or fusions with DCs) are significantly more immunogenic. These therapies are applicable to all patients, regardless of HLA-type [105]. In addition, multiple, polyclonal TAA responses have been demonstrated with long-term immunity and prevention of immune evasion [106,107]. mRNA-based vaccines have also been shown to be superior to antigenic peptide-based vaccines [108].
Novel techniques have been developed to better activate bone-marrow DCs for an anti-tumor response. Two methods using carbon black nanoparticles and antigens with mannosylated dendrimers were shown to significantly activate bone marrow DCs and increase T-cell stimulating capacity of DCs up to two fold [109,110]. Just as exosomes play a crucial role in establishing the bone metastatic niche [30]. DC-derived exosomes are capable of establishing an anti-tumoral T-cell response [111]. Multiple pre-clinical studies showed the high potency of DC-derived exosome-based therapy, including the eradication of established tumors in a mouse model [112,113]. Three phase I trials in metastatic melanoma, non-small cell lung cancer, and colorectal cancer showed high tolerability to the treatment, with only grade 1 or 2 adverse reactions. Stable disease and partial responses were observed, but due to the modest efficacy of the treatment, only one phase 2 trial is being studied [114-116]. The lower efficacy may be a limitation of the TAA used and not the exosome therapy itself. With newly identified TAAs,
14
these therapies may provide a highly effective and highly tolerated immunotherapy for bone metastases.
DCs can also be directly targeted through various treatments. These include sialidase [117], Toll-like receptor agonists (resiquimod, imiquimod, poly ICLC), and another DC-targeted adjuvant, Montanide. These treatments cause a wide range of effects including activation of lymphocytes, macrophages and DCs, while time transforming the tumor microenvironment by directly inducing apoptosis of tumor cells [118-123][124]. Clinical trials have not studied bone metastases directly, but described significant responses suggesting that DC-targeted treatments could interfere with the tumor-bone microenvironment [123,125-128].
4.3.3 CARs
CARs have also been targeted directly against the microenvironment. CARs targeted against cancer-associated fibroblasts (CAFs) present in bone metastases have seen significant efficacy. It is known that depletion of fibroblast activation protein (FAP)+ CAF reverses immunosuppression and activates T-cells [129,130]. Therefore, vaccines and CARs that target FAP have shown anti-tumor activity. Yet, when combined with vaccination or direct targeting of TAAs, the effect has been shown to be synergistic— decreasing tumor volume and increasing survival in mouse models [131-133]. Interestingly, a bispecific antibody against FAP and death receptor 5 (DR5) has shown to induce apoptosis in the non-functional cell death pathway. Most remarkable of this study was the low toxicity and complete regression of tumors when combined with irinotecan or doxorubicin in patient-derived xenograft models [134].
CARs engineered in conjunction with chemokine receptors have shown higher affinity towards the tumor microenvironment. Specifically, when chemokine (C-C motif) receptor (CCR) 2, was co-engineered with a CAR, T-cells were better trafficked to multiple types of tumor metastases [135-137]. Interestingly, the ligand for CCR2 is CCL2, which we have already shown has been implicated with macrophage recruitment and the establishment of the bone-stromal niche [86].
15
For homing specificity to prostate bone metastases, the chemokine stromal cell-derived factor (SDF)-1 also known as Chemokine (C-X-C motif) ligand (CXCL) 12 has been identified [138-140]. By engineering its receptor, chemokine C-X-C motif receptor (CXCR) 4, into CAR T cells, precise trafficking to prostatic bone metastases could be ensured [141].
Additionally, Bispecific T-cell Engagers (BiTEs) are also chimeric proteins with two scFVs to bind to CD3 on T-cells and to a surface antigen on tumor cells. BiTEs have been developed to target tumor-associated antigens in prostate cancer, melanoma, lung cancer and various other cancers [142]. BiTEs hold promise in the treating bone metastases as they are not directly affected by the immunosuppression. Although these proteins can be quickly produced and show low toxicities, they generally have less efficacy when compared to CAR therapies due to the inability to induce T-cell memory [141].
4.3.4 Improving T cell efficacy through release of TAAs
Cryoablation of skeletal metastases has been shown to bring significant pain palliation [143]. In addition to directly killing tumor tissue, cryoablation augments the immune response by the rapid release of TAAs through necrotic tumor cells [144-148]. After these responses had been observed, other studies were conducted using immune adjuvants, and subsequent improvements of tumor immunity were detected [148-150]. In these studies, complete suppression of tumor growth was observed in most mouse models. Yet, the mechanism of resistance in the few tumors that failed to respond remained unstudied until recently. The expansion of activated leukocyte cell adhesion molecule (ALCAM) positive cells was identified as the immunosuppressant mechanism in these models. When an anti-ALCAM antibody, combined with an anti-CTLA antibody and cryoablation was performed, this resistance was completely reversed, with objective responses observed in 100% of mice, and complete responses observed in 93% of mice studied [151]. These studies show that not only can cryoablation drive a significant anti-tumor response, but when resistance to these treatments
16
occurs, xenograft studies suggest that through the implementation of an anti-ALCAM antibody, this immunosuppression can almost be completely reversed.
In the case of prostate cancer, anti-androgen therapy by abiraterone and enzalutamide caused apoptotic release of TAAs and improved the sensitivity of T-cell-mediated lysis of tumor cells [152]. Radiation therapy is another suggested mechanism of antigen release to improve T-cell response [141].
Some modest improvement in immunotherapy response can be seen by increasing the number of tumor-infiltrating lymphocytes. In bone metastases, aberrant angiogenic activity promotes the proliferation of tumor cells, especially in bone metastases. Yet, due to the low quality of blood vessels, T-cells are unable to proliferate in the microenvironment [141,153]. Although anti-vascular endothelial growth factor (VEGF) treatments are meant to completely inhibit the formation of new vasculature, when used in lower doses, normalization of vasculature occurs, chemokine T-cell attractants are upregulated, and consequent T-cell infiltration to metastases occurs [153]. A synergistic reduction of tumor growth was observed when anti-angiogenic therapy was combined with ACT [154,155]. In addition, acute bacterial inflammation, in this case, directed at the prostate, increases TILs in prostate cancer [156].
Although anti-CTLA-4 and anti-PD-1 antibodies are already approved as immune checkpoint inhibitors, it has been shown that blocking TGF- and TIM-3 improves the T-cell responses. In the case of TIM-3, antibody blockade prevents exhaustion of not only T-cells but also NK-cells [102,124].
4.3.5 Oncolytic Viruses
Oncolytic viruses specifically target certain tumor cells while promoting an immune response upon cell lysis [157]. Talimogene laherparepvec, or T-Vec, is currently the only approved oncolytic virus for treatment of melanoma [158]. Vesicular stomatitis virus (VSV) is another
17
virus that is currently being studied for treatment of pancreatic cancer and leukemia [159,160]. VSV is very sensitive to IFN- response and therefore does not proliferate well in normal tissues or neoplastic tissues that do have an IFN- response[161]. One of the proposed pathways of acquired immune resistance in tumors is a defective Janis Kinase 1 (JAK1) pathway, which in turn prevents and IFN- response of antigen presentation [162]. Multiple studies in pancreatic cancer and head and neck cancer have shown that JAK 1 inhibition overcomes resistance to VSV [161,163]. Although it has not been studied, this proposed mechanism suggests that tumor cells that have acquired resistance to immune checkpoint therapies might be susceptible to infection and subsequent lysis by VSV. This provides another method to overcome resistance to immunotherapies and severe immunosuppression in the tumor-bone microenvironment
Conclusion Bone metastases are associated with significant morbidity and detrimental impact on overall outcomes of patients with several malignancies. This manuscript summarized the complexity of tumor-bone microenvironment and discussed potential therapeutic targets. Advancing the understanding of bone immune environment can inform treatment strategies including combinatorial strategies with checkpoint inhibitors.
18
Figure 1: Depiction of Bone-Tumor Immunology A) Establishment of the “Onco-niche”. B) Representation of the positive feedback mechanism of the tumor-bone vicious cycle. C) Endogenous and therapeutic anti-tumor responses. D) Pro-tumor mechanisms from the immune system.
19
Table 1. Standard of care treatment of bone metastases Treatment
SRE Treatments Zoledronic acid
denosumab
Radium-223
Immunotherapies nivolumab
Mechanism
Effect on Bone metastases
Notes
Refs
inhibition of farnesyl pyrophosphate synthase mAb against RANKL
Inhibition of Osteolysis
Action against TAMs as well Approved in adjuvant use
[164]
Localizes to bone, releases alpha radiation
Reduces osteoclast activity Cytotoxic to tumor cells by inducing dsDNA breaks
Anti-PD-1 mAb
ipilimumab
Anti-CTLA-4 mAb
pembrolizumab
Anti-PD-1 mAb
atezolizumab
Anti-PD-L1 mAB
Sipuleucel-T
Dendritic cells stimulated with GM-CSF and PAP
[164]
Offers less myelosuppression due to shorter range of alpha radiation
[52]
Currently unstudied in bone metastases Currently unstudied in bone metastases Currently unstudied in bone metastases Currently unstudied in bone metastases Unknown
[100]
20
Table 2. Emerging Targeted Therapies for Bone Metastases Treatment Mechanism Effect on Bone Outcomes metastases Tumor-targeted Osteoprotegerin Natural Inhibits Conflicting inhibitor of osteoclastogenesis outcomes RANKL and subsequent depending on bone reabsorption dose and type of administration Cabozantinib TKI, inhibits inhibition of PFS- 5.9 to 23.9 MET, VEGFR- osteoblastic and months, pain 2, RET,KIT, osteolytic lesions in palliation in 57%, AXL, FLT3 xenografts bone scan response in 63% AP12009 NucelotideReverses tumorNo bone (trabedersen) based, blocks mediated immune metastases data production suppression and reported prevented of TGF-2 metastases galunisertib Small Inhibit the boneIncreased OS, molecule tumor viscous cycle especially in inhibitor of patients with low TGF- levels of TGF- belagenpumatucel- Vaccine with Target tumor cells in OS benefits in L four TGF-b2- bone patients with prior antisense microenvironment chemotherapy or generadiotherapy that secrete TGF- modified, irradiated, allogeneic NSCLC cell lines Abiraterone + Androgen Shown to inhibit significantly prednisone synthesis progression of increase palliative inhibitor via prostatic lesions in benefit and CYP17A1 the bone decrease SREs inhibition P62 DNA Vaccine P62 in Decreases tumor Suppression of plasmid size, suppresses osteoporosis administered metastases,
Notes
Refs
New models have shown optimal dosages, need to be tested
[57,63]
OS improvement not seen in phase III trial
[75-82]
[165,166]
No specific end points measured with bone metastases No specific end points measured with bone metastases
[64,65]
Only for use in mCRPC, SoC
[168]
Currently only studied in animal models
[70-72,74]
[64,66,167]
21
created antibody response Stromal-targeted Zoledronic acid
PLX3397
Multitargeted TKI, inhibits CSF1
Bindarit
Inhibits CCL2
Tasquinimod
Blocks S100A9
Pirfenidone, and nintedanib Ac-PhScN-NH2 inhibitor
Target CAFs
Sunitinib
Imatinib
increased TIL, and decreases osteoclastogenesis
Acts on TAMs, change from M2 to M1 phenotype Prevents recruitment of M2 TAMs/MDSCs, improves immune response Prevents macrophage recruitment, Inhibits establishment of tumor-stromal niche inhibits angiogenesis, immunomodulates through TAMs, prevents of the establishment of the bone-metastatic niche
Significantly decreased tumor volume with ACT
Preclinical data only
[83,85,169]
Reduced metastasis formation
Animal models only
[86,87]
patients with bone metastases 8.8 months vs 3.4 months (placebo) PFS, no OS benefit seen
No further research after failed phase III trial
[90,91]
No data published
[36]
2nd generation is 100,000x more potent. 1st gen saw 14 months PFS Co-administered with CEA vaccine
[67,68]
51 inhibition through a small peptide TKI, blocks STAT3 and IDO pathways
prevents metastases and angiogenesis, due to vicious cycle of osteolytic lesions
reduced intratibial colony progression by almost 80 % in mouse model
Decreases MDSCs, Tregs. Increased TIL
TKI, blocks STAT3 and IDO pathways
activates CD8+ T cells, induces Treg apoptosis
Reduced tumor volume, increased OS in mouse model, No change in tumor burden in RCC patients Increased antiIn mouse models tumor response only with immunotherapies
[92,93]
[94,95]
22
Bevacizumab
Anti-VEGF mAB
Prevents dysfunction of DC into MDSCs
Temsirolimus
mTOR inhibitor, acts on IDO pathway Tetravalent FAP-DR5 Antibody
Activates CD8+ T cells, inhibits Tregs
Ex vivo DCs from MM patients functioned normally Better PFS in RCC patients
targets CAFs and defective apoptosis pathway on tumor cells
Stable disease and Low toxicities complete tumor regression with doxorubicin in vivo
[134]
Specific targeting of ACT to solid tumor DCs activated against a variety of TAAs Increase antigen uptake and activate DCs
More direct administration of ACT to bone metastases Better and more specific activation of DCs in bone microenvironment
Limited research in rat models
[170]
Significant increase of bone-marrow DC activation
Increased effectiveness of other vaccine based therapies
Endocytosis of exosomes for polyepitopic antigen presentation and response Exogenous sialic acid removal
Strong anti-tumoral T-cell response
High tolerability, with stable disease and partial responses in metastatic disease
RG7386
Immune-targeted Chitosan thermogels
Vaccines via whole tumor lysates/fusions
carbon black nanoparticles & Antigens with mannosylated dendrimers DC-derived exosomes
Sialidase
Toll-like receptor agonists (resiquimod, imiquimod, poly ICLC)
Increased maturation and stimulation of autologous T-cells Activation of Alone or in macrophages combination with DCs and vaccines, decrease other immune suppression lymphocytes in tumor
Increased survival among patients with metastatic disease
At lower doses normalization of vasculature, increased TIL
[96,153]
[97-99]
Multiple trials done, see 97
[106,107]
[109,110]
Only one phase II trial done, may be more effective with different TAA
Increased tumorcell apoptosis in murine model Dramatic results seen specifically with poly ICLC with complete tumor regression
[111-113]
[117]
Considered adjuvants, Montanide also acts similarly
[118-123]
23
microenvironment, can directly induce apoptosis of tumor cells Increase CAR T-cell proliferation and cytotoxicity Overcomes immunosuppression in tumor microenvironment
CRISPR-CaS9 edited CAR T-cells
PD-1 knockout
TRUCKs
IL-12 secreting CAR T-cells
CAR T-cells with chemokine receptors
CCR2 and CXCR4 receptors engineered into CAR Tcells Targets CAFs
Better T-cell trafficking to tumor metastases, specifically prostatic metastases for CXCR4 Reverses immunosuppression, activates T-cells against TAAs
Bispecific T-cell Engagers
Target TAA and CD3 for T-cell activation
Cryoablation
Direct destruction of metastases through liquid-cooled probes
Increased T-cell cytotoxcity in metastatic disease, unaffected by tumor immunosupression Rapid necrotic release of TAA induces immune response throughout body
Vesicular stomatitis virus
Oncolytic virus, highly sensitive to IFN- response
Anti-FAP CAR Tcells
Targets tumors with defective JAK1 pathway associated with immune resistance
No study results posted
NCT02793856
Increased efficacy of CAR T-cells, decreased immunosupression of bone-derived stroma cells Dramatic increase of TIL including in bone microenvironment
[171-173]
Synergy with vaccination, decreasing tumor volume and OS in mouse model Less efficacy compared to CARs due to inability to induce T-cell memory With anti-ALCAM antibody, complete response in 100% of mouse models, significant pain palliation in patients Currently unstudied
CXCR4 can also be [136therapeutic target, 141,174] but inhibition increases osteoclastogenesis [129-133]
Low toxicities, easily produced
[141,142]
Use of Anti-CTLA antibody and adjuvants also improves response
[144150,175]
Could be next-line treatment after failure of immunotherapies
[158-163]
24
References: [1] [2] [3]
[4]
[5] [6] [7]
[8]
[9]
[10] [11] [12] [13] [14] [15] [16] [17] [18]
Weilbaecher, K. N., Guise, T. A., and McCauley, L. K., 2011, “Cancer to bone: a fatal attraction,” arXiv, 11(6), pp. 411–425. Coleman, R. E., 2006, “Clinical features of metastatic bone disease and risk of skeletal morbidity,” Clinical Cancer Research, 12(20 Pt 2), pp. 6243s–6249s. Clemons, M., Gelmon, K. A., Pritchard, K. I., and Paterson, A. H., 2012, “Bone-targeted agents and skeletal-related events in breast cancer patients with bone metastases: the state of the art,” Curr Oncol, 19(5), pp. 259–268. Zustovich, F., and Fabiani, F., 2014, “Therapeutic opportunities for castration-resistant prostate cancer patients with bone metastases,” Critical Reviews in Oncology / Hematology, 91(2), pp. 197–209. Fidler, I. J., 2003, “The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited,” arXiv, 3(6), pp. 453–458. Wan, L., Pantel, K., and Kang, Y., 2013, “Tumor metastasis: moving new biological insights into the clinic,” Nature Medicine, 19(11), pp. 1450–1464. Braun, S., Kentenich, C., Janni, W., Hepp, F., de Waal, J., Willgeroth, F., Sommer, H., and Pantel, K., 2000, “Lack of effect of adjuvant chemotherapy on the elimination of single dormant tumor cells in bone marrow of high-risk breast cancer patients,” Journal of Clinical Oncology, 18(1), pp. 80–86. Mohme, M., Riethdorf, S., and Pantel, K., 2017, “Circulating and disseminated tumour cells - mechanisms of immune surveillance and escape.,” Nat Rev Clin Oncol, 14(3), pp. 155–167. Pantel, K., and Alix-Panabières, C., 2014, “Bone marrow as a reservoir for disseminated tumor cells: a special source for liquid biopsy in cancer patients.,” BoneKEy reports, 3, p. 584. Kang, Y., and Pantel, K., 2013, “Tumor Cell Dissemination: Emerging Biological Insights from Animal Models and Cancer Patients,” Cancer Cell, 23(5), pp. 573–581. Rucci, N., and Teti, A., 2010, “Osteomimicry: how tumor cells try to deceive the bone,” Front Biosci (Schol Ed), 2, pp. 907–915. Jadaan, D. Y., Jadaan, M. M., and McCabe, J. P., 2015, “Cellular Plasticity in Prostate Cancer Bone Metastasis.,” Prostate Cancer, 2015(18), pp. 651580–12. Kan, C., Vargas, G., Le Pape, F., and Clezardin, P., 2016, “Cancer Cell Colonisation in the Bone Microenvironment,” International journal of molecular sciences, 17(10). Bussard, K. M., Gay, C. V., and Mastro, A. M., 2008, “The bone microenvironment in metastasis; what is special about bone?,” Cancer Metastasis Rev., 27(1), pp. 41–55. Ortiz, A., and Lin, S. H., 2012, “Osteolytic and osteoblastic bone metastases: two extremes of the same spectrum?,” Recent Results Cancer Res, 192, pp. 225–233. Matsumoto, T., and Abe, M., 2011, “TGF-beta-related mechanisms of bone destruction in multiple myeloma,” Bone, 48(1), pp. 129–134. Westendorf, J. J., Kahler, R. A., and Schroeder, T. M., 2004, “Wnt signaling in osteoblasts and bone diseases,” Gene, 341, pp. 19–39. Xiao, G., Jiang, D., Ge, C., Zhao, Z., Lai, Y., Boules, H., Phimphilai, M., Yang, X., 25
[19]
[20] [21] [22] [23]
[24] [25]
[26]
[27] [28] [29] [30]
[31] [32]
Karsenty, G., and Franceschi, R. T., 2005, “Cooperative interactions between activating transcription factor 4 and Runx2/Cbfa1 stimulate osteoblast-specific osteocalcin gene expression,” J Biol Chem, 280(35), pp. 30689–30696. Li, J., Sarosi, I., Yan, X. Q., Morony, S., Capparelli, C., Tan, H. L., McCabe, S., Elliott, R., Scully, S., Van, G., Kaufman, S., Juan, S. C., Sun, Y., Tarpley, J., Martin, L., Christensen, K., McCabe, J., Kostenuik, P., Hsu, H., Fletcher, F., Dunstan, C. R., Lacey, D. L., and Boyle, W. J., 2000, “RANK is the intrinsic hematopoietic cell surface receptor that controls osteoclastogenesis and regulation of bone mass and calcium metabolism,” Proc. Natl. Acad. Sci. U.S.A., 97(4), pp. 1566–1571. Soki, F. N., Park, S. I., and McCauley, L. K., 2012, “The multifaceted actions of PTHrP in skeletal metastasis,” Future Oncol, 8(7), pp. 803–817. Mundy, G. R., 2002, “Metastasis to bone: causes, consequences and therapeutic opportunities,” arXiv, 2(8), pp. 584–593. Capietto, A. H., and Faccio, R., 2014, “Immune regulation of bone metastasis,” BoneKEy reports, 3, p. 600. D'Amico, L., and Roato, I., 2015, “The Impact of Immune System in Regulating Bone Metastasis Formation by Osteotropic Tumors,” Journal of Immunology Research, 2015, p. 143526. Foss, F. M., 2002, “Immunologic mechanisms of antitumor activity,” YSONC, 29(3 Suppl 7), pp. 5–11. Kunzmann, V., Kimmel, B., Herrmann, T., Einsele, H., and Wilhelm, M., 2009, “Inhibition of phosphoantigen-mediated gammadelta T-cell proliferation by CD4+ CD25+ FoxP3+ regulatory T cells,” Immunology, 126(2), pp. 256–267. Monteiro, A. C., Leal, A. C., Goncalves-Silva, T., Mercadante, A. C., Kestelman, F., Chaves, S. B., Azevedo, R. B., Monteiro, J. P., and Bonomo, A., 2013, “T cells induce pre-metastatic osteolytic disease and help bone metastases establishment in a mouse model of metastatic breast cancer,” PloS one, 8(7), p. e68171. Gabrilovich, D. I., Ostrand-Rosenberg, S., and Bronte, V., 2012, “Coordinated regulation of myeloid cells by tumours,” Nat Rev Immunol, 12(4), pp. 253–268. Vasiliadou, I., and Holen, I., 2013, “The role of macrophages in bone metastasis,” Journal of Bone Oncology, 2(4), pp. 158–166. Croucher, P. I., McDonald, M. M., and Martin, T. J., 2016, “Bone metastasis: the importance of the neighbourhood.,” arXiv, 16(6), pp. 373–386. Peinado, H., Aleckovic, M., Lavotshkin, S., Matei, I., Costa-Silva, B., Moreno-Bueno, G., Hergueta-Redondo, M., Williams, C., Garcia-Santos, G., Ghajar, C. M., NitadoriHoshino, A., Hoffman, C., Badal, K., Garcia, B. A., Callahan, M. K., Yuan, J., Martins, V. R., Skog, J., Kaplan, R. N., Brady, M. S., Wolchok, J. D., Chapman, P. B., Kang, Y., Bromberg, J., and Lyden, D., 2012, “Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET,” Nature Medicine, 18(6), pp. 883–. Baschuk, N., Rautela, J., and Parker, B. S., 2015, “Bone specific immunity and its impact on metastasis,” BoneKEy reports, 4, p. 665. Zhao, E., Xu, H., Wang, L., Kryczek, I., Wu, K., Hu, Y., Wang, G., and Zou, W., 2012, “Bone marrow and the control of immunity,” Cellular & molecular immunology, 9(1), 26
[33]
[34] [35]
[36]
[37]
[38]
[39]
[40]
[41]
[42]
[43] [44]
[45]
pp. 11–19. Topalian, S. L., Drake, C. G., and Pardoll, D. M., 2012, “Targeting the PD-1/B7-H1(PDL1) pathway to activate anti-tumor immunity,” Curr Opin Immunol, 24(2), pp. 207– 212. Hodi, F. S., 2010, “Overcoming immunological tolerance to melanoma: Targeting CTLA-4,” Asia Pac J Clin Oncol, 6 Suppl 1, pp. S16–23. Sakuishi, K., Apetoh, L., Sullivan, J. M., Blazar, B. R., Kuchroo, V. K., and Anderson, A. C., 2010, “Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity,” J. Exp. Med., 207(10), pp. 2187–2194. Antonia, S. J., Vansteenkiste, J. F., and Moon, E., 2016, “Immunotherapy: Beyond Anti–PD-1 and Anti–PD-L1 Therapies,” American Society of Clinical Oncology Educational Book, 36, pp. e450–e458. Zarek, P. E., Huang, C. T., Lutz, E. R., Kowalski, J., Horton, M. R., Linden, J., Drake, C. G., and Powell, J. D., 2008, “A2A receptor signaling promotes peripheral tolerance by inducing T-cell anergy and the generation of adaptive regulatory T cells,” Blood, 111(1), pp. 251–259. Bidwell, B. N., Slaney, C. Y., Withana, N. P., Forster, S., Cao, Y., Loi, S., Andrews, D., Mikeska, T., Mangan, N. E., Samarajiwa, S. A., de Weerd, N. A., Gould, J., Argani, P., ller, A. M. O., Smyth, M. J., Anderson, R. L., Hertzog, P. J., and Parker, B. S., 2012, “Silencing of Irf 7 pathways in breast cancer cells promotes bone metastasis through immune escape,” Nature Medicine, 18(8), pp. 1224–1231. Malladi, S., Macalinao, D. G., Jin, X., He, L., Basnet, H., Zou, Y., de Stanchina, E., and Massagué, J., 2016, “Metastatic Latency and Immune Evasion through Autocrine Inhibition of WNT,” Cell, 165(1), pp. 45–60. Bottos, A., Gotthardt, D., Gill, J. W., Gattelli, A., Frei, A., Tzankov, A., Sexl, V., WodnarFilipowicz, A., and Hynes, N. E., 2016, “Decreased NK-cell tumour immunosurveillance consequent to JAK inhibition enhances metastasis in breast cancer models,” Nat Commun, 7, p. 12258. Amo, L., Tamayo-Orbegozo, E., Maruri, N., Eguizabal, C., Zenarruzabeitia, O., Rinon, M., Arrieta, A., Santos, S., Monge, J., Vesga, M. A., Borrego, F., and Larrucea, S., 2014, “Involvement of platelet-tumor cell interaction in immune evasion. Potential role of podocalyxin-like protein 1,” Front Oncol, 4, p. 245. Criscitiello, C., Viale, G., Gelao, L., Esposito, A., De Laurentiis, M., De Placido, S., Santangelo, M., Goldhirsch, A., and Curigliano, G., 2015, “Crosstalk between bone niche and immune system: osteoimmunology signaling as a potential target for cancer treatment,” CANCER TREATMENT REVIEWS, 41(2), pp. 61–68. Rogers, M. J., Watts, D. J., and Russell, R., 1997, “Overview of bisphosphonates,” Cancer, 80(8), pp. 1652–1660. Dougall, W. C., Glaccum, M., Charrier, K., Rohrbach, K., Brasel, K., De Smedt, T., Daro, E., Smith, J., Tometsko, M. E., Maliszewski, C. R., Armstrong, A., Shen, V., Bain, S., Cosman, D., Anderson, D., Morrissey, P. J., Peschon, J. J., and Schuh, J., 1999, “RANK is essential for osteoclast and lymph node development,” Genes & Development, 13(18), pp. 2412–2424. Castellano, D., Sepulveda, J. M., Garcia-Escobar, I., Rodriguez-Antolin, A., Sundlov, A., 27
[46]
[47]
[48]
[49]
[50]
[51]
[52] [53]
and Cortes-Funes, H., 2011, “The Role of RANK-Ligand Inhibition in Cancer: The Story of Denosumab,” The Oncologist, 16(2), pp. 136–145. Himelstein, A. L., Qin, R., Novotny, P. J., Seisler, D. K., Khatcheressian, J. L., Roberts, J. D., Grubbs, S. S., O'Connor, T., Weckstein, D., Loprinzi, C. L., and Shapiro, C. L., 2017, “CALGB 70604 (Alliance): A randomized phase III study of standard dosing vs. longer interval dosing of zoledronic acid in metastatic cancer.,” Journal of Clinical Oncology. Biermann, J. S., Chow, W., Reed, D. R., Lucas, D., Adkins, D. R., Agulnik, M., Benjamin, R. S., Brigman, B., Budd, G. T., Curry, W. T., Didwania, A., Fabbri, N., Hornicek, F. J., Kuechle, J. B., Lindskog, D., Mayerson, J., McGarry, S. V., Million, L., Morris, C. D., Movva, S., O'Donnell, R. J., Randall, R. L., Rose, P., Santana, V. M., Satcher, R. L., Schwartz, H., Siegel, H. J., Thornton, K., Villalobos, V., Bergman, M. A., and Scavone, J. L., 2017, “NCCN Guidelines Insights: Bone Cancer, Version 2.2017.,” J Natl Compr Canc Netw, 15(2), pp. 155–167. Stopeck, A. T., Lipton, A., Body, J. J., Steger, G. G., Tonkin, K., de Boer, R. H., Lichinitser, M., Fujiwara, Y., Yardley, D. A., Viniegra, M., Fan, M., Jiang, Q., Dansey, R., Jun, S., and Braun, A., 2010, “Denosumab Compared With Zoledronic Acid for the Treatment of Bone Metastases in Patients With Advanced Breast Cancer: A Randomized, Double-Blind Study,” Journal of Clinical Oncology, 28(35), pp. 5132– 5139. Gnant, M., Pfeiler, G., Dubsky, P. C., Hubalek, M., Greil, R., Jakesz, R., Wette, V., Balic, M., Haslbauer, F., Melbinger, E., Bjelic-Radisic, V., Artner-Matuschek, S., Fitzal, F., Marth, C., Sevelda, P., Mlineritsch, B., Steger, G. G., Manfreda, D., Exner, R., Egle, D., Bergh, J., Kainberger, F., Talbot, S., Warner, D., Fesl, C., Singer, C. F., Austrian Breast and Colorectal Cancer Study Group, 2015, “Adjuvant denosumab in breast cancer (ABCSG-18): a multicentre, randomised, double-blind, placebo-controlled trial.,” Lancet, 386(9992), pp. 433–443. Krop, I., Johnston, S., Mayer, I. A., Dickler, M., Ganju, V., Forero-Torres, A., Melichar, B., Morales, S., de Boer, R., Gendreau, S., Derynck, M., Lackner, M., Spoerke, J., Yeh, R.-F., Levy, G., Ng, V., O'Brien, C., Savage, H., Xiao, Y., Wilson, T., Lee, S. C., Petrakova, K., Vallentin, S., Yardley, D., Ellis, M., Piccart, M., Perez, E. A., Winer, E., and Schmid, P., 2015, “Abstract S2-02: The FERGI phase II study of the PI3K inhibitor pictilisib (GDC-0941) plus fulvestrant vs fulvestrant plus placebo in patients with ER+, aromatase inhibitor (AI)-resistant advanced or metastatic breast cancer – Part I results,” Cancer Research, 75(9 Supplement), pp. S2–02–S2–02. Early Breast Cancer Trialists' Collaborative Group (EBCTCG), Coleman, R., Powles, T., Paterson, A., Gnant, M., Anderson, S., Diel, I., Gralow, J., Minckwitz, von, G., Moebus, V., Bergh, J., Pritchard, K. I., Bliss, J., Cameron, D., Evans, V., Pan, H., Peto, R., Bradley, R., and Gray, R., 2015, “Adjuvant bisphosphonate treatment in early breast cancer: meta-analyses of individual patient data from randomised trials.,” Lancet, 386(10001), pp. 1353–1361. Aragon-Ching, and El-Amm, J., 2016, “Targeting Bone Metastases in Metastatic Castration-Resistant Prostate Cancer,” CMO, pp. 11–9. Parker, C., Nilsson, S., Heinrich, D., O'Sullivan, J. M., Fossa, S. D., Chodacki, A., Wiechno, P. J., Logue, J. P., Seke, M., Widmark, A., Johannessen, D. C., Hoskin, P., 28
[54]
[55]
[56] [57]
[58] [59]
[60]
[61]
[62]
[63] [64] [65]
[66]
Bottomley, D., Coleman, R. E., Vogelzang, N. J., O'Bryan-Tear, C. G., Garcia-Vargas, J. E., Shan, M., and Sartor, A. O., 2012, “Updated analysis of the phase III, double-blind, randomized, multinational study of radium-223 chloride in castration-resistant prostate cancer (CRPC) patients with bone metastases (ALSYMPCA).,” Journal of Clinical Oncology, 30(15). Fahmy, O., Khairul-Asri, M. G., Stenzl, A., and Gakis, G., 2016, “The current status of checkpoint inhibitors in metastatic bladder cancer,” Clinical & Experimental Metastasis, pp. 1–7. Ratta, R., Zappasodi, R., Raggi, D., Grassi, P., Verzoni, E., Necchi, A., Di Nicola, M., Salvioni, R., de Braud, F., and Procopio, G., 2016, “Immunotherapy advances in urogenital malignancies,” Critical Reviews in Oncology / Hematology, pp. 1–13. Rogers, T. L., and Holen, I., 2011, “Tumour macrophages as potential targets of bisphosphonates,” pp. 1–17. Morony, S., Capparelli, C., Sarosi, I., Lacey, D. L., Dunstan, C. R., and Kostenuik, P. J., 2001, “Osteoprotegerin inhibits osteolysis and decreases skeletal tumor burden in syngeneic and nude mouse models of experimental bone metastasis,” Cancer Research, 61(11), pp. 4432–4436. Corey, E., 2005, “Osteoprotegerin in Prostate Cancer Bone Metastasis,” Cancer Research, 65(5), pp. 1710–1718. Fisher, J. L., Thomas-Mudge, R. J., Elliott, J., Hards, D. K., Sims, N. A., Slavin, J., Martin, T. J., and Gillespie, M. T., 2006, “Osteoprotegerin overexpression by breast cancer cells enhances orthotopic and osseous tumor growth and contrasts with that delivered therapeutically,” Cancer Research, 66(7), pp. 3620–3628. Brown, J. M., Vessella, R. L., Kostenuik, P. J., Dunstan, C. R., Lange, P. H., and Corey, E., 2001, “Serum osteoprotegerin levels are increased in patients with advanced prostate cancer,” Clinical Cancer Research, 7(10), pp. 2977–2983. Chen, G., Sircar, K., Aprikian, A., Potti, A., Goltzman, D., and Rabbani, S. A., 2006, “Expression of RANKL/RANK/OPG in primary and metastatic human prostate cancer as markers of disease stage and functional regulation,” Cancer, 107(2), pp. 289–298. Chamoux, E., Houde, N., L'eriger, K., and Roux, S., 2008, “Osteoprotegerin decreases human osteoclast apoptosis by inhibiting the TRAIL pathway,” J. Cell. Physiol., 216(2), pp. 536–542. Ryser, M. D., Qu, Y., and Komarova, S. V., 2012, “Osteoprotegerin in bone metastases: mathematical solution to the puzzle.,” PLoS Comput Biol, 8(10), p. e1002703. de Gramont, A., Faivre, S., and Raymond, E., 2017, “Novel TGF-β inhibitors ready for prime time in onco-immunology,” OncoImmunology, 6(1), pp. 1–5. Melisi, D., Garcia-Carbonero, R., Macarulla, T., Pezet, D., Deplanque, G., Fuchs, M., Trojan, J., Oettle, H., Kozloff, M., Cleverly, A., Gueorguieva, I., Desaiah, D., Lahn, M. M., Blunt, Al, Benhadji, K. A., and Tabernero, J., 2016, “Abstract CT068: A randomized phase II, double-blind study to evaluate the efficacy and safety of galunisertib+gemcitabine (GG) or gemcitabine+placebo (GP) in patients with unresectable pancreatic cancer (PC),” Cancer Research, 76(14 Supplement), pp. CT068–CT068. Nemunaitis, J., and Giaccone, G., 2014, “A Phase III Study of Belagenpumatucel-L, an 29
[67]
[68]
[69]
[70]
[71]
[72] [73]
[74]
[75]
[76]
[77]
Allogeneic Tumor Cell Vaccine, as Maintenance Therapy for Non-Small Cell Lung Cancer,” Molecular Therapy, 22, p. S200. Jia, Y., Zeng, Z.-Z., Markwart, S. M., Rockwood, K. F., Ignatoski, K. M. W., Ethier, S. P., and Livant, D. L., 2004, “Integrin fibronectin receptors in matrix metalloproteinase-1dependent invasion by breast cancer and mammary epithelial cells.,” Cancer Research, 64(23), pp. 8674–8681. Yao, H., Veine, D. M., and Livant, D. L., 2016, “Therapeutic inhibition of breast cancer bone metastasis progression and lung colonization: breaking the vicious cycle by targeting α5β1 integrin.,” Breast cancer research and treatment, 157(3), pp. 489–501. Cianfrocca, M. E., Kimmel, K. A., Gallo, J., Cardoso, T., Brown, M. M., Hudes, G., Lewis, N., Weiner, L., Lam, G. N., Brown, S. C., Shaw, D. E., Mazar, A. P., and Cohen, R. B., 2006, “Phase 1 trial of the antiangiogenic peptide ATN-161 (Ac-PHSCN-NH2), a beta integrin antagonist, in patients with solid tumours,” British Journal of Cancer, 94(11), pp. 1621–1626. Durán, A., Serrano, M., Leitges, M., Flores, J. M., Picard, S., Brown, J. P., Moscat, J., and Diaz-Meco, M. T., 2004, “The atypical PKC-interacting protein p62 is an important mediator of RANK-activated osteoclastogenesis.,” Dev. Cell, 6(2), pp. 303–309. Sabbieti, M. G., Agas, D., Capitani, M., Marchetti, L., Concetti, A., Vullo, C., Catone, G., Gabai, V., Shifrin, V., Sherman, M. Y., Shneider, A., and Venanzi, F. M., 2015, “Plasmid DNA-coding p62 as a bone effective anti-inflammatory/anabolic agent.,” Oncotarget, 6(6), pp. 3590–3599. Zhang, J., Yang, Z., and Dong, J., 2016, “P62: An emerging oncotarget for osteolytic metastasis,” Journal of Bone Oncology, 5(1), pp. 30–37. Venanzi, F., Shifrin, V., Sherman, M., Gabai, V., Kiselev, O., Komissarov, A., Grudinin, M., Shartukova, M., Romanovskaya-Romanko, E. A., Kudryavets, Y., Bezdenezhnykh, N., Lykhova, O., Semesyuk, N., Concetti, A., Tsyb, A., Filimonova, M., Makarchuk, V., Yakubovsky, R., Chursov, A., Shcherbinina, V., and Shneider, A., 2013, “Broadspectrum anti-tumor and anti-metastatic DNA vaccine based on p62-encoding vector.,” Oncotarget, 4(10), pp. 1829–1835. Gabai, V., Venanzi, F. M., Bagashova, E., Rud, O., Mariotti, F., Vullo, C., Catone, G., Sherman, M. Y., Concetti, A., Chursov, A., Latanova, A., Shcherbinina, V., Shifrin, V., and Shneider, A., 2014, “Pilot study of p62 DNA vaccine in dogs with mammary tumors.,” Oncotarget, 5(24), pp. 12803–12810. Yakes, F. M., Chen, J., Tan, J., Yamaguchi, K., Shi, Y., Yu, P., Qian, F., Chu, F., Bentzien, F., Cancilla, B., Orf, J., You, A., Laird, A. D., Engst, S., Lee, L., Lesch, J., Chou, Y.-C., and Joly, A. H., 2011, “Cabozantinib (XL184), a novel MET and VEGFR2 inhibitor, simultaneously suppresses metastasis, angiogenesis, and tumor growth.,” Mol. Cancer Ther., 10(12), pp. 2298–2308. Schimmoller, F., Zayzafoon, M., Chung, L. W. K., Zhau, H. E., Fagerlund, K. M., and Aftab, D. T., 2011, “Abstract A233: Cabozantinib (XL184), a dual MET-VEGFR2 inhibitor, blocks osteoblastic and osteolytic progression of human prostate cancer xenografts in mouse bone.,” Mol. Cancer Ther., 10(Supplement 1), pp. A233–A233. Smith, M. R., Sweeney, C. J., Corn, P. G., Rathkopf, D. E., Smith, D. C., Hussain, M., George, D. J., Higano, C. S., Harzstark, A. L., Sartor, A. O., Vogelzang, N. J., Gordon, M. 30
[78]
[79]
[80]
[81]
[82]
[83]
[84]
S., De Bono, J. S., Haas, N. B., Logothetis, C. J., Elfiky, A., Scheffold, C., Laird, A. D., Schimmoller, F., Basch, E. M., and Scher, H. I., 2014, “Cabozantinib in ChemotherapyPretreated Metastatic Castration-Resistant Prostate Cancer: Results of a Phase II Nonrandomized Expansion Study,” Journal of Clinical Oncology, 32(30), pp. 3391– 3399. Smith, D. C., Smith, M. R., Sweeney, C., Elfiky, A. A., Logothetis, C., Corn, P. G., Vogelzang, N. J., Small, E. J., Harzstark, A. L., Gordon, M. S., Vaishampayan, U. N., Haas, N. B., Spira, A. I., Lara, P. N., Lin, C.-C., Srinivas, S., Sella, A., Schöffski, P., Scheffold, C., Weitzman, A. L., and Hussain, M., 2013, “Cabozantinib in patients with advanced prostate cancer: results of a phase II randomized discontinuation trial.,” J. Clin. Oncol., 31(4), pp. 412–419. Smith, M. R., De Bono, J. S., Sternberg, C. N., Le Moulec, S., Oudard, S., De Giorgi, U., Krainer, M., Bergman, A. M., Hoelzer, W., De Wit, R., Boegemann, M., Saad, F., Cruciani, G., Thiery- Vuillemin, A., Feyerabend, S., Miller, K., Ramies, D. A., Hessel, C., Weitzman, A., and Fizazi, K., 2015, “Final analysis of COMET-1: Cabozantinib (Cabo) versus prednisone (Pred) in metastatic castration-resistant prostate cancer (mCRPC) patients (pts) previously treated with docetaxel (D) and abiraterone (A) and/or enzalutamide (E).,” ASCO Meeting Abstracts, 33(7_suppl), p. 139. Basch, E. M., Scholz, M. C., De Bono, J. S., Vogelzang, N. J., De Souza, P. L., Marx, G. M., Vaishampayan, U. N., George, S., Schwarz, J. K., Antonarakis, E. S., O'Sullivan, J. M., Rezazadeh Kalebasty, A., Chi, K. N., Dreicer, R., Hutson, T. E., Mangeshkar, M., Holland, J. S., Weitzman, A., and Scher, H. I., 2015, “Final analysis of COMET-2: Cabozantinib (Cabo) versus mitoxantrone/prednisone (MP) in metastatic castrationresistant prostate cancer (mCRPC) patients (pts) with moderate to severe pain who were previously treated with docetaxel (D) and abiraterone (A) and/or enzalutamide (E).,” ASCO Meeting Abstracts, 33(7_suppl), p. 141. Choueiri, T. K., Escudier, B., Powles, T., Tannir, N. M., Mainwaring, P. N., Rini, B. I., Hammers, H. J., Donskov, F., Roth, B. J., Peltola, K., Lee, J. L., Heng, D. Y. C., Schmidinger, M., Agarwal, N., Sternberg, C. N., McDermott, D. F., Aftab, D. T., Hessel, C., Scheffold, C., Schwab, G., Hutson, T. E., Pal, S., Motzer, R. J., METEOR investigators, 2016, “Cabozantinib versus everolimus in advanced renal cell carcinoma (METEOR): final results from a randomised, open-label, phase 3 trial.,” Lancet Oncol., 17(7), pp. 917–927. Escudier, B. J., Motzer, R. J., Powles, T., Tannir, N. M., Davis, I. D., Donskov, F., Grunwald, V., Heng, D. Y. C., Hutson, T., Melichar, B., Nosov, D., Rini, B. I., Salman, P., Sternberg, C. N., Szczylik, C., Wolter, P., Arroyo, A. M., Mangeshkar, M., Agarwal, N., and Choueiri, T. K., 2016, “Subgroup analyses of METEOR, a randomized phase 3 trial of cabozantinib versus everolimus in patients (pts) with advanced renal cell carcinoma (RCC).,” ASCO Meeting Abstracts, 34(2_suppl), p. 499. Mok, S., Koya, R. C., Tsui, C., Xu, J., Robert, L., Wu, L., Graeber, T. G., West, B. L., Bollag, G., and Ribas, A., 2014, “Inhibition of CSF-1 Receptor Improves the Antitumor Efficacy of Adoptive Cell Transfer Immunotherapy,” Cancer Research, 74(1), pp. 153– 161. Ngiow, S. F., Meeth, K. M., Stannard, K., Barkauskas, D. S., Bollag, G., Bosenberg, M., 31
[85]
[86]
[87]
[88]
[89]
[90]
[91]
[92]
[93]
[94]
and Smyth, M. J., 2015, “Co-inhibition of colony stimulating factor-1 receptor and BRAF oncogene in mouse models of BRAFV600E melanoma,” OncoImmunology, 5(3), p. e1089381. Sluijter, M., van der Sluis, T. C., van der Velden, P. A., Versluis, M., West, B. L., van der Burg, S. H., and van Hall, T., 2014, “Inhibition of CSF-1R supports T-cell mediated melanoma therapy.,” PloS one, 9(8), p. e104230. Steiner, J. L., Davis, J. M., McClellan, J. L., Guglielmotti, A., and Murphy, E. A., 2014, “Effects of the MCP-1 synthesis inhibitor bindarit on tumorigenesis and inflammatory markers in the C3(1)/SV40Tag mouse model of breast cancer,” Cytokine, 66(1), pp. 60–68. Zollo, M., Di Dato, V., Spano, D., De Martino, D., Liguori, L., Marino, N., Vastolo, V., Navas, L., Garrone, B., Mangano, G., Biondi, G., and Guglielmotti, A., 2012, “Targeting monocyte chemotactic protein-1 synthesis with bindarit induces tumor regression in prostate and breast cancer animal models,” Clinical & Experimental Metastasis, 29(6), pp. 585–601. Gupta, N., Ustwani, Al, O., Shen, L., and Pili, R., 2014, “Mechanism of action and clinical activity of tasquinimod in castrate-resistant prostate cancer,” Onco Targets Ther, 7, pp. 223–234. Raymond, E., Dalgleish, A., Damber, J. E., Smith, M., and Pili, R., 2014, “Mechanisms of action of tasquinimod on the tumour microenvironment,” Cancer Chemother. Pharmacol., 73(1), pp. 1–8. Pili, R., Häggman, M., Stadler, W. M., Gingrich, J. R., Assikis, V. J., Bjork, A., Nordle, O., Forsberg, G., Carducci, M. A., and Armstrong, A. J., 2011, “Phase II Randomized, Double-Blind, Placebo-Controlled Study of Tasquinimod in Men With Minimally Symptomatic Metastatic Castrate-Resistant Prostate Cancer,” Journal of Clinical Oncology, 29(30), pp. 4022–4028. Sternberg, C., Armstrong, A., Pili, R., Ng, S., Huddart, R., Agarwal, N., Khvorostenko, D., Lyulko, O., Brize, A., Vogelzang, N., Delva, R., Harza, M., Thanos, A., James, N., Werbrouck, P., Bogemann, M., Hutson, T., Milecki, P., Chowdhury, S., Gallardo, E., Schwartsmann, G., Pouget, J. C., Baton, F., Nederman, T., Tuvesson, H., and Carducci, M., 2016, “Randomized, Double-Blind, Placebo-Controlled Phase III Study of Tasquinimod in Men With Metastatic Castration-Resistant Prostate Cancer,” Journal of Clinical Oncology, pp. 1–16. Farsaci, B., Higgins, J. P., and Hodge, J. W., 2012, “Consequence of dose scheduling of sunitinib on host immune response elements and vaccine combination therapy,” Int. J. Cancer, 130(8), pp. 1948–1959. Ko, J. S., Zea, A. H., Rin, B. I., Ireland, J. L., Elson, P., Cohen, P., Golshayan, A., Rayman, P. A., Wood, L., Garcia, J., Dreicer, R., Bukowski, R., and Finke, J. H., 2009, “Sunitinib Mediates Reversal of Myeloid-Derived Suppressor Cell Accumulation in Renal Cell Carcinoma Patients,” Clinical Cancer Research, 15(6), pp. 2148–2157. Balachandran, V. P., Cavnar, M. J., Zeng, S., Bamboat, Z. M., Ocuin, L. M., Obaid, H., Sorenson, E. C., Popow, R., Ariyan, C., Rossi, F., Besmer, P., Guo, T., Antonescu, C. R., Taguchi, T., Yuan, J., Wolchok, J. D., Allison, J. P., and DeMatteo, R. P., 2011, “Imatinib potentiates antitumor T cell responses in gastrointestinal stromal tumor through the 32
[95]
[96]
[97]
[98]
[99] [100] [101]
[102]
[103]
[104]
[105]
[106]
inhibition of Ido,” Nature Medicine, 17(9), pp. 1094–U99. Larmonier, N., Janikashvili, N., LaCasse, C. J., Larmonier, C. B., Cantrell, J., Situ, E., Lundeen, T., Bonnotte, B., and Katsanis, E., 2008, “Imatinib mesylate inhibits CD4+ CD25+ regulatory T cell activity and enhances active immunotherapy against BCRABL- tumors.,” J Immunol, 181(10), pp. 6955–6963. Yang, D.-H., Park, J.-S., Jin, C.-J., Kang, H.-K., Nam, J.-H., Rhee, J.-H., Kim, Y.-K., Chung, S.-Y., Choi, S.-J.-N., Kim, H.-J., Chung, I.-J., and Lee, J.-J., 2009, “The dysfunction and abnormal signaling pathway of dendritic cells loaded by tumor antigen can be overcome by neutralizing VEGF in multiple myeloma,” Leukemia Research, 33(5), pp. 665–670. Procaccini, C., De Rosa, V., Galgani, M., Abanni, L., Cali, G., Porcellini, A., Carbone, F., Fontana, S., Horvath, T. L., La Cava, A., and Matarese, G., 2010, “An Oscillatory Switch in mTOR Kinase Activity Sets Regulatory T Cell Responsiveness,” Immunity, 33(6), pp. 929–941. Wang, Y., Camirand, G., Lin, Y., Froicu, M., Deng, S., Shlomchik, W. D., Lakkis, F. G., and Rothstein, D. M., 2011, “Regulatory T Cells Require Mammalian Target of Rapamycin Signaling To Maintain Both Homeostasis and Alloantigen-Driven Proliferation in Lymphocyte-Replete Mice,” J Immunol, 186(5), pp. 2809–2818. Chen, S.-C., and Kuo, P.-L., 2016, “Bone Metastasis from Renal Cell Carcinoma,” International journal of molecular sciences, 17(6), pp. 987–918. Vanneman, M., and Dranoff, G., 2012, “Combining immunotherapy and targeted therapies in cancer treatment,” arXiv, 12(4), pp. 237–251. Restifo, N. P., Dudley, M. E., and Rosenberg, S. A., 2012, “Adoptive immunotherapy for cancer: harnessing the T cell response,” Nature Reviews Immunology, 12(4), pp. 269–281. Baitsch, L., Baumgaertner, P., Devêvre, E., Raghav, S. K., Legat, A., Barba, L., Wieckowski, S., Bouzourene, H., Deplancke, B., Romero, P., Rufer, N., and Speiser, D. E., 2011, “Exhaustion of tumor-specific CD8⁺ T cells in metastases from melanoma patients.,” The Journal of Clinical Investigation, 121(6), pp. 2350–2360. Rosenberg, S. A., Yang, J. C., Sherry, R. M., Kammula, U. S., Hughes, M. S., Phan, G. Q., Citrin, D. E., Restifo, N. P., Robbins, P. F., Wunderlich, J. R., Morton, K. E., Laurencot, C. M., Steinberg, S. M., White, D. E., and Dudley, M. E., 2011, “Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy.,” Clinical Cancer Research, 17(13), pp. 4550–4557. Robbins, P. F., Morgan, R. A., Feldman, S. A., Yang, J. C., Sherry, R. M., Dudley, M. E., Wunderlich, J. R., Nahvi, A. V., Helman, L. J., Mackall, C. L., Kammula, U. S., Hughes, M. S., Restifo, N. P., Raffeld, M., Lee, C.-C. R., Levy, C. L., Li, Y. F., El-Gamil, M., Schwarz, S. L., Laurencot, C., and Rosenberg, S. A., 2011, “Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1.,” J. Clin. Oncol., 29(7), pp. 917–924. Kajihara, M., Takakura, K., Kanai, T., Ito, Z., Saito, K., Takami, S., Shimodaira, S., Okamoto, M., Ohkusa, T., and Koido, S., 2016, “Dendritic cell-based cancer immunotherapy for colorectal cancer,” WJG, 22(17), pp. 4275–13. de Gruijl, T. D., van den Eertwegh, A. J. M., Pinedo, H. M., and Scheper, R. J., 2008, 33
[107]
[108] [109]
[110]
[111]
[112] [113]
[114]
[115]
[116]
[117]
[118]
“Whole-cell cancer vaccination: from autologous to allogeneic tumor- and dendritic cell-based vaccines.,” Cancer Immunol. Immunother., 57(10), pp. 1569–1577. Kajihara, M., Takakura, K., Ohkusa, T., and Koido, S., 2015, “The impact of dendritic cell-tumor fusion cells on cancer vaccines - past progress and future strategies.,” Immunotherapy, 7(10), pp. 1111–1122. Gilboa, E., and Vieweg, J., 2004, “Cancer immunotherapy with mRNA‐transfected dendritic cells,” Immunol. Rev., 199(1), pp. 251–263. Koike, E., Takano, H., Inoue, K.-I., Yanagisawa, R., and Kobayashi, T., 2008, “Carbon black nanoparticles promote the maturation and function of mouse bone marrowderived dendritic cells.,” Chemosphere, 73(3), pp. 371–376. Sheng, K.-C., Kalkanidis, M., Pouniotis, D. S., Esparon, S., Tang, C. K., Apostolopoulos, V., and Pietersz, G. A., 2008, “Delivery of antigen using a novel mannosylated dendrimer potentiates immunogenicity in vitro and in vivo.,” Eur. J. Immunol., 38(2), pp. 424–436. Klippstein, R., and Pozo, D., 2010, “Nanotechnology-based manipulation of dendritic cells for enhanced immunotherapy strategies,” Nanomedicine: Nanotechnology, Biology, and Medicine, 6(4), pp. 523–529. Amigorena, S., 2000, “Cancer immunotherapy using dendritic cell-derived exosomes.,” Medicina (B Aires), 60 Suppl 2, pp. 51–54. Zitvogel, L., Regnault, A., Lozier, A., Wolfers, J., Flament, C., Tenza, D., RicciardiCastagnoli, P., Raposo, G., and Amigorena, S., 1998, “Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes.,” Nature Medicine, 4(5), pp. 594–600. Dai, S., Wei, D., Wu, Z., Zhou, X., Wei, X., Huang, H., and Li, G., 2008, “Phase I Clinical Trial of Autologous Ascites-derived Exosomes Combined With GM-CSF for Colorectal Cancer,” Molecular Therapy, 16(4), pp. 782–790. Morse, M. A., Garst, J., Osada, T., Khan, S., Hobeika, A., Clay, T. M., Valente, N., Shreeniwas, R., Sutton, M. A., Delcayre, A., Hsu, D.-H., Le Pecq, J.-B., and Lyerly, H. K., 2005, “A phase I study of dexosome immunotherapy in patients with advanced nonsmall cell lung cancer,” J Transl Med, 3(1), p. 1. Escudier, B., Dorval, T., Chaput, N., André, F., Caby, M.-P., Novault, S., Flament, C., Leboulaire, C., Borg, C., Amigorena, S., Boccaccio, C., Bonnerot, C., Dhellin, O., Movassagh, M., Piperno, S., Robert, C., Serra, V., Valente, N., Le Pecq, J.-B., Spatz, A., Lantz, O., Tursz, T., Angevin, E., and Zitvogel, L., 2005, “Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: results of thefirst phase I clinical trial.,” J Transl Med, 3(1), p. 10. Silva, M., Silva, Z., Marques, G., Ferro, T., Gonçalves, M., Monteiro, M., van Vliet, S. J., Mohr, E., Lino, A. C., Fernandes, A. R., Lima, F. A., van Kooyk, Y., Matos, T., Tadokoro, C. E., and Videira, P. A., 2016, “Sialic acid removal from dendritic cells improves antigen cross-presentation and boosts anti-tumor immune responses,” Oncotarget, pp. 1–14. Liu, K., Idoyaga, J., Charalambous, A., Fujii, S.-I., Bonito, A., Mordoh, J., Wainstok, R., Bai, X.-F., Liu, Y., and Steinman, R. M., 2005, “Innate NKT lymphocytes confer superior adaptive immunity via tumor-capturing dendritic cells.,” J. Exp. Med., 202(11), pp. 34
[119]
[120]
[121]
[122]
[123]
[124]
[125]
[126]
[127]
1507–1516. Thapa, P., Zhang, G., Xia, C., Gelbard, A., Overwijk, W. W., Liu, C., Hwu, P., Chang, D. Z., Courtney, A., Sastry, J. K., Wang, P. G., Li, C., and Zhou, D., 2009, “Nanoparticle formulated alpha-galactosylceramide activates NKT cells without inducing anergy.,” Vaccine, 27(25-26), pp. 3484–3488. Wang, J., Cho, S., Ueno, A., Cheng, L., Xu, B.-Y., Xu, B.-Y., Desrosiers, M. D., Shi, Y., and Yang, Y., 2008, “Ligand-dependent induction of noninflammatory dendritic cells by anergic invariant NKT cells minimizes autoimmune inflammation.,” J Immunol, 181(4), pp. 2438–2445. Valmori, D., Souleimanian, N. E., Tosello, V., Bhardwaj, N., Adams, S., O'Neill, D., Pavlick, A., Escalon, J. B., Cruz, C. M., Angiulli, A., Angiulli, F., Mears, G., Vogel, S. M., Pan, L., Jungbluth, A. A., Hoffmann, E. W., Venhaus, R., Ritter, G., Old, L. J., and Ayyoub, M., 2007, “Vaccination with NY-ESO-1 protein and CpG in Montanide induces integrated antibody/Th1 responses and CD8 T cells through cross-priming.,” Proc. Natl. Acad. Sci. U.S.A., 104(21), pp. 8947–8952. Sabado, R. L., Pavlick, A., Gnjatic, S., Cruz, C. M., Vengco, I., Hasan, F., Spadaccia, M., Darvishian, F., Chiriboga, L., Holman, R. M., Escalon, J., Muren, C., Escano, C., Yepes, E., Sharpe, D., Vasilakos, J. P., Rolnitzsky, L., Goldberg, J. D., Mandeli, J., Adams, S., Jungbluth, A., Pan, L., Venhaus, R., Ott, P. A., and Bhardwaj, N., 2015, “Resiquimod as an immunologic adjuvant for NY-ESO-1 protein vaccination in patients with high-risk melanoma.,” Cancer Immunol Res, 3(3), pp. 278–287. Salazar, A. M., Erlich, R. B., Mark, A., Bhardwaj, N., and Herberman, R. B., 2014, “Therapeutic In Situ Autovaccination against Solid Cancers with Intratumoral PolyICLC: Case Report, Hypothesis, and Clinical Trial,” Cancer Immunol Res, 2(8), pp. 720– 724. Gonzalez-Gugel, E., Saxena, M., and Bhardwaj, N., 2016, “Modulation of innate immunity in the tumor microenvironment,” Cancer Immunology, Immunotherapy, pp. 1–8. Rapoport, A. P., Aqui, N. A., Stadtmauer, E. A., Vogl, D. T., Xu, Y. Y., Kalos, M., Cai, L., Fang, H.-B., Weiss, B. M., Badros, A., Yanovich, S., Akpek, G., Tsao, P., Cross, A., Mann, D., Philip, S., Kerr, N., Brennan, A., Zheng, Z., Ruehle, K., Milliron, T., Strome, S. E., Salazar, A. M., Levine, B. L., and June, C. H., 2014, “Combination immunotherapy after ASCT for multiple myeloma using MAGE-A3/Poly-ICLC immunizations followed by adoptive transfer of vaccine-primed and costimulated autologous T cells.,” Clinical Cancer Research, 20(5), pp. 1355–1365. Okada, H., Butterfield, L. H., Hamilton, R. L., Hoji, A., Sakaki, M., Ahn, B. J., Kohanbash, G., Drappatz, J., Engh, J., Amankulor, N., Lively, M. O., Chan, M. D., Salazar, A. M., Shaw, E. G., Potter, D. M., and Lieberman, F. S., 2015, “Induction of robust type-I CD8+ T-cell responses in WHO grade 2 low-grade glioma patients receiving peptidebased vaccines in combination with poly-ICLC.,” Clinical Cancer Research, 21(2), pp. 286–294. Pollack, I. F., Jakacki, R. I., Butterfield, L. H., Hamilton, R. L., Panigrahy, A., Normolle, D. P., Connelly, A. K., Dibridge, S., Mason, G., Whiteside, T. L., and Okada, H., 2016, “Immune responses and outcome after vaccination with glioma-associated antigen 35
[128]
[129]
[130]
[131]
[132]
[133]
[134]
[135]
[136]
[137]
peptides and poly-ICLC in a pilot study for pediatric recurrent low-grade gliomas,” Neuro Oncol, 18(8), pp. 1157–1168. Hartman, L. L. R., Crawford, J. R., Makale, M. T., Milburn, M., Joshi, S., Salazar, A. M., Hasenauer, B., VandenBerg, S. R., MacDonald, T. J., and Durden, D. L., 2014, “Pediatric Phase II Trials of Poly-ICLC in the Management of Newly Diagnosed and Recurrent Brain Tumors,” Journal of Pediatric Hematology/Oncology, 36(6), pp. 451–457. Kraman, M., Bambrough, P. J., Arnold, J. N., Roberts, E. W., Magiera, L., Jones, J. O., Gopinathan, A., Tuveson, D. A., and Fearon, D. T., 2010, “Suppression of Antitumor Immunity by Stromal Cells Expressing Fibroblast Activation Protein- ,” Science, 330(6005), pp. 827–830. Zhang, Y., and Ertl, H. C. J., 2016, “Depletion of FAP+ cells reduces immunosuppressive cells and improves metabolism and functions CD8+T cells within tumors,” Oncotarget, pp. 1–18. Wang, L.-C. S., Lo, A., Scholler, J., Sun, J., Majumdar, R. S., Kapoor, V., Antzis, M., Cotner, C. E., Johnson, L. A., Durham, A. C., Solomides, C. C., June, C. H., Pure, E., and Albelda, S. M., 2014, “Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe toxicity.,” Cancer Immunol Res, 2(2), pp. 154–166. Kakarla, S., Chow, K. K. H., Mata, M., Shaffer, D. R., Song, X.-T., Wu, M.-F., Liu, H., Wang, L. L., Rowley, D. R., Pfizenmaier, K., and Gottschalk, S., 2013, “Antitumor effects of chimeric receptor engineered human T cells directed to tumor stroma.,” Mol. Ther., 21(8), pp. 1611–1620. Gottschalk, S., Yu, F., Ji, M., Kakarla, S., and Song, X.-T., 2013, “A Vaccine That CoTargets Tumor Cells and Cancer Associated Fibroblasts Results in Enhanced Antitumor Activity by Inducing Antigen Spreading,” PloS one, 8(12), p. e82658. Brünker, P., Wartha, K., Friess, T., Grau-Richards, S., Waldhauer, I., Koller, C. F., Weiser, B., Majety, M., Runza, V., Niu, H., Packman, K., Feng, N., Daouti, S., Hosse, R. J., Mössner, E., Weber, T. G., Herting, F., Scheuer, W., Sade, H., Shao, C., Liu, B., Wang, P., Xu, G., Vega-Harring, S., Klein, C., Bosslet, K., and Umaña, P., 2016, “RG7386, a Novel Tetravalent FAP-DR5 Antibody, Effectively Triggers FAP-Dependent, AvidityDriven DR5 Hyperclustering and Tumor Cell Apoptosis.,” Mol. Cancer Ther., 15(5), pp. 946–957. Moon, E. K., Carpenito, C., Sun, J., Wang, L.-C. S., Kapoor, V., Predina, J., Powell, D. J., Riley, J. L., June, C. H., and Albelda, S. M., 2011, “Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelin-specific chimeric antibody receptor.,” Clinical Cancer Research, 17(14), pp. 4719–4730. Asai, H., Fujiwara, H., An, J., Ochi, T., Miyazaki, Y., Nagai, K., Okamoto, S., Mineno, J., Kuzushima, K., Shiku, H., Inoue, H., and Yasukawa, M., 2013, “Co-Introduced Functional CCR2 Potentiates In Vivo Anti-Lung Cancer Functionality Mediated by T Cells Double Gene-Modified to Express WT1-Specific T-Cell Receptor,” PloS one, 8(2), p. e56820. Craddock, J. A., Lu, A., Bear, A., Pule, M., Brenner, M. K., Rooney, C. M., and Foster, A. E., 2010, “Enhanced Tumor Trafficking of GD2 Chimeric Antigen Receptor T Cells by 36
[138]
[139]
[140]
[141] [142] [143]
[144]
[145]
[146]
[147]
[148]
[149]
Expression of the Chemokine Receptor CCR2b,” Journal of Immunotherapy, 33(8), pp. 780–788. Bleul, C. C., Fuhlbrigge, R. C., Casasnovas, J. M., Aiuti, A., and Springer, T. A., 1996, “A highly efficacious lymphocyte chemoattractant, stromal cell-derived factor 1 (SDF-1),” J. Exp. Med., 184(3), pp. 1101–1109. Kantele, J. M., Kurk, S., and Jutila, M. A., 2000, “Effects of continuous exposure to stromal cell-derived factor-1 alpha on T cell rolling and tight adhesion to monolayers of activated endothelial cells.,” J Immunol, 164(10), pp. 5035–5040. Hirbe, A. C., Morgan, E. A., and Weilbaecher, K. N., 2010, “The CXCR4/SDF-1 Chemokine Axis: A Potential Therapeutic Target for Bone Metastases?,” Current Pharmaceutical Design, 16(11), pp. 1284–1290. Hillerdal, V., and Essand, M., 2015, “Chimeric Antigen Receptor-Engineered T Cells for the Treatment of Metastatic Prostate Cancer,” BioDrugs, pp. 1–15. Fan, G., Wang, Z., Hao, M., and Li, J., 2015, “Bispecific antibodies and their applications,” Journal of Hematology & Oncology, pp. 1–14. Callstrom, M. R., Dupuy, D. E., Solomon, S. B., Beres, R. A., Littrup, P. J., Davis, K. W., Paz-Fumagalli, R., Hoffman, C., Atwell, T. D., Charboneau, J. W., Schmit, G. D., Goetz, M. P., Rubin, J., Brown, K. J., Novotny, P. J., and Sloan, J. A., 2012, “Percutaneous image-guided cryoablation of painful metastases involving bone,” Cancer, 119(5), pp. 1033–1041. Michael S. Sabel, 2009, “Cryo-immunology: A review of the literature and proposed mechanisms for stimulatory versus suppressive immune responses,” Cryobiology, 58(1), pp. 1–11. Gazzaniga, S., Bravo, A., Goldszmid, S. R., Maschi, F., Martinelli, J., Mordoh, J., and Wainstok, R., 2001, “Inflammatory Changes after Cryosurgery-Induced Necrosis in Human Melanoma Xenografted in Nude Mice,” Journal of Investigative Dermatology, 116(5), pp. 664–671. Ravindranath, M. H., Wood, T. F., Soh, D., Gonzales, A., Muthugounder, S., Perez, C., Morton, D. L., and Bilchik, A. J., 2002, “Cryosurgical ablation of liver tumors in colon cancer patients increases the serum total ganglioside level and then selectively augments antiganglioside IgM.,” Cryobiology, 45(1), pp. 10–21. Brok, den, M. H. M. G. M., Sutmuller, R. P. M., Nierkens, S., Bennink, E. J., Frielink, C., Toonen, L. W. J., Boerman, O. C., Figdor, C. G., Ruers, T. J. M., and Adema, G. J., 2006, “Efficient loading of dendritic cells following cryo and radiofrequency ablation in combination with immune modulation induces anti-tumour immunity,” British Journal of Cancer, 95(7), pp. 896–905. Udagawa, M., Kudo-Saito, C., Hasegawa, G., Yano, K., Yamamoto, A., Yaguchi, M., Toda, M., Azuma, I., Iwai, T., and Kawakami, Y., 2006, “Enhancement of Immunologic Tumor Regression by Intratumoral Administration of Dendritic Cells in Combination with Cryoablative Tumor Pretreatment and Bacillus Calmette-Guerin Cell Wall Skeleton Stimulation,” Clinical Cancer Research, 12(24), pp. 7465–7475. Brok, den, M. H. M. G. M., 2006, “Synergy between In situ Cryoablation and TLR9 Stimulation Results in a Highly Effective In vivo Dendritic Cell Vaccine,” Cancer Research, 66(14), pp. 7285–7292. 37
[150]
[151]
[152]
[153]
[154]
[155]
[156]
[157] [158] [159]
[160]
[161]
[162]
Waitz, R., Solomon, S. B., Petre, E. N., Trumble, A. E., Fasso, M., Norton, L., and Allison, J. P., 2012, “Potent Induction of Tumor Immunity by Combining Tumor Cryoablation with Anti-CTLA-4 Therapy,” Cancer Research, 72(2), pp. 430–439. Kudo-Saito, C., Fuwa, T., and Kawakami, Y., 2016, “Targeting ALCAM in the cryotreated tumour microenvironment successfully induces systemic anti-tumour immunity,” arXiv, 62(C), pp. 54–61. Ardiani, A., Gameiro, S. R., Kwilas, A. R., Donahue, R. N., and Hodge, J. W., 2014, “Androgen deprivation therapy sensitizes prostate cancer cells to T-cell killing through androgen receptor dependent modulation of the apoptotic pathway.,” Oncotarget, 5(19), pp. 9335–9348. Huang, H., Langenkamp, E., Georganaki, M., Loskog, A., Fuchs, P. F., Dieterich, L. C., Kreuger, J., and Dimberg, A., 2015, “VEGF suppresses T-lymphocyte infiltration in the tumor microenvironment through inhibition of NF-κB-induced endothelial activation.,” FASEB J., 29(1), pp. 227–238. Shi, S., Wang, R., Chen, Y., Song, H., Chen, L., and Huang, G., 2013, “Combining antiangiogenic therapy with adoptive cell immunotherapy exerts better antitumor effects in non-small cell lung cancer models.,” PloS one, 8(6), p. e65757. Shi, S., Chen, L., and Huang, G., 2013, “Antiangiogenic therapy improves the antitumor effect of adoptive cell immunotherapy by normalizing tumor vasculature.,” Med. Oncol., 30(4), pp. 698–7. Simons, B. W., Durham, N. M., Bruno, T. C., Grosso, J. F., Schaeffer, A. J., Ross, A. E., Hurley, P. J., Berman, D. M., Drake, C. G., Thumbikat, P., and Schaeffer, E. M., 2015, “A human prostatic bacterial isolate alters the prostatic microenvironment and accelerates prostate cancer progression,” J. Pathol., 235(3), pp. 478–489. Russell, S. J., Peng, K.-W., and Bell, J. C., 2012, “Oncolytic virotherapy,” Nature Biotechnology, 30(7), pp. 658–670. Greig, S. L., 2016, “Talimogene Laherparepvec: First Global Approval,” Drugs, 76(1), pp. 147–154. Zhou, Y., Wen, F., Zhang, P., Tang, R., and Li, Q., 2016, “Vesicular stomatitis virus is a potent agent for the treatment of malignant ascites.,” Oncol. Rep., 35(3), pp. 1573– 1581. Felt, S. A., Moerdyk-Schauwecker, M. J., and Grdzelishvili, V. Z., 2015, “Induction of apoptosis in pancreatic cancer cells by vesicular stomatitis virus.,” Virology, 474, pp. 163–173. Escobar-Zarate, D., Liu, Y.-P., Suksanpaisan, L., Russell, S. J., and Peng, K.-W., 2013, “Overcoming cancer cell resistance to VSV oncolysis with JAK1/2 inhibitors,” Cancer Gene Ther, 20(10), pp. 582–589. Zaretsky, J. M., Garcia-Diaz, A., Shin, D. S., Escuin-Ordinas, H., Hugo, W., Hu-Lieskovan, S., Torrejon, D. Y., Abril-Rodriguez, G., Sandoval, S., Barthly, L., Saco, J., Homet Moreno, B., Mezzadra, R., Chmielowski, B., Ruchalski, K., Shintaku, I. P., Sanchez, P. J., Puig-Saus, C., Cherry, G., Seja, E., Kong, X., Pang, J., Berent-Maoz, B., Comin-Anduix, B., Graeber, T. G., Tumeh, P. C., Schumacher, T. N. M., Lo, R. S., and Ribas, A., 2016, “Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma,” N Engl J Med, pp. NEJMoa1604958–11. 38
[163]
[164]
[165]
[166]
[167]
[168]
[169]
[170]
[171]
[172]
Cataldi, M., Shah, N. R., Felt, S. A., and Grdzelishvili, V. Z., 2015, “Breaking resistance of pancreatic cancer cells to an attenuated vesicular stomatitis virus through a novel activity of IKK inhibitor TPCA-1,” Virology, 485(C), pp. 340–354. Vignani, F., Bertaglia, V., Buttigliero, C., Tucci, M., Scagliotti, G. V., and Di Maio, M., 2016, “Skeletal metastases and impact of anticancer and bone-targeted agents in patients with castration-resistant prostate cancer,” CANCER TREATMENT REVIEWS, 44(C), pp. 61–73. Achyut, B. R., and Yang, L., 2011, “Transforming Growth Factor-β in the Gastrointestinal and Hepatic Tumor Microenvironment,” Gastroenterology, 141(4), pp. 1167–1178. Hau, P., Jachimczak, P., Schlingensiepen, R., Schulmeyer, F., Jauch, T., Steinbrecher, A., Brawanski, A., Proescholdt, M., Schlaier, J., Buchroithner, J., Pichler, J., Wurm, G., Mehdorn, M., Strege, R., Schuierer, G., Villarrubia, V., Fellner, F., Jansen, O., Straube, T., Nohria, V., Goldbrunner, M., Kunst, M., Schmaus, S., Stauder, G., Bogdahn, U., and Schlingensiepen, K.-H., 2007, “Inhibition of TGF-beta 2 with AP 12009 in recurrent malignant gliomas: From preclinical to phase I/II studies,” Oligonucleotides, 17(2), pp. 201–212. Nemunaitis, J., Dillman, R. O., Schwarzenberger, P. O., Senzer, N., Cunningham, C., Cutler, J., Tong, A., Kumar, P., Pappen, B., Hamilton, C., DeVol, E., Maples, P. B., Liu, L., Chamberlin, T., Shawler, D. L., and Fakhrai, H., 2006, “Phase II study of belagenpumatucel-L, a transforming growth factor beta-2 antisense gene-modified allogeneic tumor cell vaccine in non-small-cell lung cancer.,” J. Clin. Oncol., 24(29), pp. 4721–4730. Logothetis, C. J., Basch, E., MD, A. M., MD, P. K. F., MD, S. A. N., MD, K. N. C., MD, R. J. J., MD, O. B. G., MD, P. N. M., MD, C. N. S., MD, E. E., MA, D. D. G., PhD, M. R., PhD, Y. H., PhD, C. S. L., PhD, T. S. K., MD, C. M. H., MD, P. H. I. S., and de Bono MD, P. J. S., 2012, “Effect of abiraterone acetate and prednisone compared with placebo and prednisone on pain control and skeletal-related events in patients with metastatic castration-resistant prostate cancer: exploratory analysis of data from the COU-AA301 randomised trial,” Lancet Oncology, 13(12), pp. 1210–1217. Ngiow, S. F., Meeth, K. M., Stannard, K., Barkauskas, D. S., Bollag, G., Bosenberg, M., and Smyth, M. J., 2016, “Co-inhibition of colony stimulating factor-1 receptor and BRAF oncogene in mouse models of BRAF(V600E) melanoma.,” OncoImmunology, 5(3), p. e1089381. Monette, A., Ceccaldi, C., Assaad, E., Lerouge, S., and Lapointe, R., 2016, “Chitosan thermogels for local expansion and delivery of tumor-specific T lymphocytes towards enhanced cancer immunotherapies,” Biomaterials, 75(C), pp. 237–249. Chmielewski, M., Hombach, A. A., and Abken, H., 2014, “Of CARs and TRUCKs: chimeric antigen receptor (CAR) T cells engineered with an inducible cytokine to modulate the tumor stroma,” Immunol. Rev., 257(1), pp. 83–90. Chmielewski, M., and Abken, H., 2012, “CAR T cells transform to trucks: chimeric antigen receptor–redirected T cells engineered to deliver inducible IL-12 modulate the tumour stroma to combat cancer,” Cancer Immunology, Immunotherapy, 61(8), pp. 1269–1277. 39
[173]
[174]
[175]
Kerkar, S. P., Goldszmid, R. S., Muranski, P., Chinnasamy, D., Yu, Z., Reger, R. N., Leonardi, A. J., Morgan, R. A., Wang, E., Marincola, F. M., Trinchieri, G., Rosenberg, S. A., and Restifo, N. P., 2011, “IL-12 triggers a programmatic change in dysfunctional myeloid-derived cells within mouse tumors,” J. Clin. Invest., 121(12), pp. 4746–4757. Moon, E. K., Ranganathan, R., Eruslanov, E., Kim, S., Newick, K., O'Brien, S., Lo, A., Liu, X., Zhao, Y., and Albelda, S. M., 2016, “Blockade of Programmed Death 1 Augments the Ability of Human T Cells Engineered to Target NY-ESO-1 to Control Tumor Growth after Adoptive Transfer,” Clinical Cancer Research, 22(2), pp. 436–447. Kudo-Saito, C., Fuwa, T., and Kawakami, Y., 2016, “Targeting ALCAM in the cryotreated tumour microenvironment successfully induces systemic anti-tumour immunity,” arXiv, 62(C), pp. 54–61.
40