phase i/iib trial: in-situ vaccine for metastatic colorectal cancer · 2018-01-21 · cd8 as idc...

1
CD8 As iDC engulfs HSP, it matures to DC 1 Dendritic Cell Maturation: The HSP are engulfed by iDC which process the alloantigens chaperoned on the released HSP. In the presence of DAMP, the iDC mature to IL-12+ DC1. These activated DC1 traffic to the draining lymph nodes, upregulate CD80/86 co-stimulatory molecules and present the processed alloantigens on MHCI and MHCII. Initiation: In the priming phase, there are multiple intradermal (ID) injections of BAG cells. GM-CSF produced by the BAG, attracts NK cells and immature DC (iDC), such as Langerhans’s cells, to the injection site. NK cells in the presence of IFN-γ upregulate NKG2D-L expression enabling recognition and lysis of the allogeneic NKG2D+ BAG cells. Lysing of the BAG cells causes release of endogenous DAMP and HSP chaperones of the alloantigens into the microenvironment, creating an in-situ anti-alloantigen vaccine. iDC engulf and process the alloantigens to initiate the anti-alloantigen immune cascade. Once allo-specific immunity is established, subsequent ID BAG injections can also be rejected by the allo-specific Th1/CTL. Non-Specific Lysis of Tumor Cells The extravasating waves of activated NK cells and activated Type 1 memory cells encounter a tumor microenvironment that is profoundly immunosuppressive, with resident myeloid-derived suppressor cells (MDSC), Treg, Th2/Tr1, tumor-associated macrophages (TAM) and cancer-associated fibroblasts (CAF). These suppressor cells maintain a Type 2 cytokine environment dominated by IL-10, TGF-β and IL-6 which suppresses cellular immune function. In a Type 2 environment, tumor cells down-regulate MHC I and co-stimulatory molecule expression and upregulate expression of checkpoint molecules (e.g., CTLA4/PD-L1) all which inhibit recognition and attack by CTL. Infiltrating activated NK cells and Type 1 memory cells condition the tumor microenvironment by contributing Type 1 cytokines such as IFN-γ , TNF-α and IL-2. These Type 1 cytokines cause differentiation of resident macrophages to the M1 tumoricidal phenotype, counter-regulate the effects of infiltrating suppressor cells, and upregulate counter death receptors, MHC I and co-stimulatory molecules on tumor cells. Under these conditions, non-specific tumor lysis can occur through activated NK cells, M1 macrophages and activated memory cells. Tumor lysis releases endogenous DAMP and endogenous tumor neoantigens chaperoned on HSP. The released HSP are engulfed and processed by iDC, which in the presence of DAMP and Type 1 cytokines mature to IL-12+ DC1 and traffic to the draining lymph node to initiate the tumor-specific immune cascade. Tumor Bloodstream Activation Once in circulation, the activated, non-memory, allo-specific Th1 and CTL T-cells produce IFN-γ and express CD40L. This causes activation of circulating NK cells and memory T-cells. Activated NK cells and memory cells traffic to the tumor sites. Activated memory cells express ‘death receptors’ such as FasL, TRAIL and TWEEK which can non-specifically kill tumors in a Type 1 cytokine environment. Subsequent BAG injections release additional waves of activated, non-memory, allo-specific Th1 and CTL T-cells into circulation which, in turn, causes new waves of activated NK cells and memory cells to infiltrate tumor sites. After each BAG injection, a portion of the activated, non-memory, allo-specific Th1 and CTL will differentiate into memory cells. As the titer of circulating memory allo-specific Th1 and CTL increases in circulation, each wave of activated, non-memory, allo-specific Th1 and CTL will non-specifically activate increasing numbers of allo-specific Th1 and CTL cells from the circulating memory pool . As increased numbers of activated NK and Th1/CTL memory cells enter the tumor microenvironment, the production of Type 1 cytokines modulates the tumor environment from Type 2 to Type 1 dominance. Priming Phase In the priming phase, BAG cells are injected intradermally in order to elicit high titers of allo-specific Th1/CTL in circulation. These activated T-cells in turn non-specifically activate circulating NK cells, M1 macrophages and memory T-cells through production of IFN-γ and CD40:CD40L interaction. All these non-specific effector cells traffic to tumor sites and condition the local microenvironment with Type 1 cytokines In the presence of Type 1 cy- tokines, all these non-specifically activated cells can lyse tumor cells. This tumor lysis results in the release of endogenous heat shock proteins (HSP) and danger-associated molecular patterns (DAMP) from the lysed tumors. HSP chaperone the complete tumor antigen repertoire, including neoantigens. Therefore, released HSP in the context of the Type 1 cytokine environment and DAMP, provides an in-situ vaccine with tumor neoantigen and ad- juvant danger signals for eliciting tumor-specific immunity. Vaccination Phase The vaccination phase serves to increase the infiltration and effector function of tumor antigen specific Th1/CTL within the tumor lesions and development of tumor-specific memory. Each priming injection of BAG cells causes waves of activated non-memory allospecific Th1/CTL and memory allospecific Th1/CTL. Each wave of non-memory allospecific Th1/CTL causes activation and extravasation of memory allospecific Th1/CTL and NK cells which conditions the tumor mi- croenvironment with Type 1 cytokines. In this environment, NK cells and activated memory allo-specific Th1/CTL along with resident M1 macrophages mediate non-specific tumor lysis. The non-specific lysis of tumor cells releases endogenous chaperoned tumor neoantigens and DAMP. The presence of tumor neoantigens and DAMP in a Type 1 environment creates an in-situ anti-tumor vaccine. iDC process the tumor antigens, traffic to lymph nodes and cause release of activated non-memory tumor-specific Th1/CTL in circulation. These cells activate circulating memory cells the same as activated non-memory allo-specific cells. As additional waves of tumor lysis occur, a portion of the activated non-memory tumor-specific Th1/CTL differentiate to memory cells. As the titers of both allo-specific and tumor-specific memory cells increase in cir- culation, each wave of activated non-memory allo-specific and tumor specific cells activates increased numbers of allo-specific and tumor-specific memory cells. DC 1 migrates to draining lymph node DC 1 migrates to draining lymph node Alloantigen Chaperoned on HSP Tumor antigen Chaperoned on HSP i DC Allo-Specific CTL and TH1 T-Cell Activation: In the draining lymph nodes, the DC1 interact with naïve CD4+ and CD8+ T-cells and produce IL-12. In the presence of IL-12, naïve CD4+ and CD8+ T-cells, which recognize the alloantigens, differentiate to allo-specific Th1 and CTL, respectively. These activated, allo-specific T-cells produce IFN-γ , upregulate CD40L and down-regulate CD62L. Down-regulation of CD62L permits their entry into the circulation. DC 1 MHC I molecule MHC II molecule Co-stimulatory molecule CD 80/86 Education signals are received as naïve T-Cells interact with their cognate antigens on the MHC I and MHC II molecules Arming signals are received via CD80/86 molecules on the DC1 to CD28 surface molecules on the naïve T-Cells Education signals are received as naïve T-Cells interact with their cognate antigens on the MHC I and MHC II molecules Arming signals are received via CD80/86 molecules on the DC1 to CD28 surface molecules on the naïve T-Cells Allo-Specific Immunity Allo-Specific Immunity Tumor-Specific Immunity Tumor-Specific Immunity Tumor-Specific Immunity Innate Immunity LYMPH NODE DC 1 LYMPH NODE DC 1 Adaptive Immunity Adaptive Immunity i DC As iDC engulfs HSP, it matures to DC 1 iDC engulfs HSP and matures to DC 1 iDC engulfs HSP and matures to DC 1 iDC engulfs HSP and matures to DC 1 DC 1 MHC I MHC II Amplification The in-situ vaccine effect caused by the non-specific lysis of tumor cells in a Type 1 cytokine envi- ronment, produces new waves of activated non-memory tumor-specific Th1/CTL. As this cas- cade feeds forward, a proportion of the non-memory tumor-specific Th1/CTL differentiate to memory cells. Each new wave of activated non-memory allo-specific and/or non-memory tu- mor-specific Th1/CTL activates these circulating memory cells causing them to traffic to tumor lesions. To amplify these effects, a select tumor lesion is killed through use of a percutaneous cryoablation followed immediately by injection of BAG cells. The cryoablation freeze-thaw cycles cause massive release of endogenous DAMP and HSP chaperones from lysed tumor cells. The insertion of the allogeneic BAG cells into the lesion recruits allo-specific Th1/CTL memory cells to reject the BAG, releasing additional DAMP and Type 1 cytokines. The immune stimulato- ry effects of the alloantigens mixed with the tumor antigens in the context of Type 1 cytokines and DAMP, amplifies the development of tumor-specific memory Th1/CTL. Activation and Booster The priming and vaccination phases serve to increase the titers of allo-specific and tumor-specif- ic Th1/CTL memory cells. BAG produce IFN-ϒ and express high density CD40L. Intravenous infu- sion of BAG increases the extravasation of these memory cells to tumor lesions through CD40L:CD40 interaction in a Type 1 cytokine environment. The allo-specific rejection response to BAG in circulation serves to release endogenous DAMP which activates circulating monocytes causing a Type 1 “cytokine storm”. The Type 1 cytokines serve to counter-regulate immunosup- pressive and immunoavoidance mechanisms and support on-going anti-tumor cellular immune responses. In the event that the Type 1 cytokine storm wanes between dosing and prior to com- plete elimination of tumor, the anti-tumor effects of can be re-activated through subsequent BAG intravenous booster infusions. In the event of over-activation of the immune system, resulting in immune-mediated toxicity, steroids can be administered. Since dexamethasone causes apoptosis of activated Th1/CTL, it may serve as a “reversal” agent. CD80/86 DC 1 migrates to draining lymph node DC 1 migrates to draining lymph node Adaptive Immunity i DC DC 1 i DC DC 1 Adaptive Immunity DC 1 migrates to draining lymph node i DC DC 1 Bioengineered Allogeneic Graft (BAG) Cells: Intentionally-mismatched, ex-vivo expanded and differ- entiated memory CD4+ Th1 cells with anti-CD3/CD28 microbeads attached that express high density CD40L, MHC II, NKG2D and Type 1 cytokines including IFN-γ, TNF-α and GM-CSF. Percutaneous Cryoablation Tumor Allogeneic BAG Cell Injection into Lesion Adaptive Immunity Counter-regulate Immunosuppressive and Immunoavoidance Th2 Mechanisms Adaptive Immunity NK NK CD4 Discussion This novel therapeutic vaccine platform is being tested for ability to elicit immune-mediated anti-tu- mor effects in third-line MSI-Low metastatic CRC pa- tients not responsive to checkpoint blockade immu- notherapy. The mechanism of the vaccine acts to pro- vide feed-forward amplifying waves of innate anti-tu- mor effects which transition to waves of tumor-specif- ic immune effects. The mechanism also incorporates a strategy for counter-regulating the immunosuppres- sive tumor microenvironment. Some cohorts incorpo- rate an in-situ vaccination strategy that combines per- cutaneous cryoablation of a tumor lesion followed by intratumoral injection of BAG cells. In-situ vaccination provides additional amplification of the anti-tumor effects of the vaccine. In order to evaluate anti-tumor responses and distinguish pseudoprogression from actual progression, the protocol evaluates longitudi- nal simultaneous CT scans and biopsy samples of a select tumor lesion. Peripheral blood samples are also collected for evaluation of various biomarkers and immune parameters. Phase I/IIb Trial: In-Situ Vaccine for Metastatic Colorectal Cancer Abstract Background: Except for MSI-H tumors, colorectal cancer (CRC) does not respond to immunotherapy. CRC does respond to the graft vs. tumor (GVT) immune effect that occurs after allo- geneic stem cell transplantation (ASCT). GVT is associated with graft vs. host disease (GVHD) toxicity which limits the clinical application. A bioengineered allograft (BAG) has been developed which can elicit host-mediated GVT-like effects without GVHD toxicity, chemotherapy condition- ing or a HLA-matched donor. BAG are Th1 memory cells derived from blood of healthy donors with CD3/CD28 mi- crobeads attached. These cells have immunomodulatory properties which enable modulation of Type 1/Type 2 bal- ance and dysregulation of immunosuppressive circuits. We are evaluating the safety and efficacy of BAG in third-line metastatic CRC. Methods: The study uses a standard 3+3 design followed by an ex- pansion phase with the optimal dosing pattern. The pro- tocol has four components: (A) priming; (B) in-situ vacci- nation; (C) activation; and (D) booster. Priming involves intradermal injections of BAG cells which activates NK cells and develops allo-specific Th1/CTL immunity. In-situ vaccination involves tumor cryoablation to release en- dogenous heat shock proteins (HSP) which chaperone tu- mor-specific neoantigens, followed immediately by the intralesional injection of BAG cells as adjuvant. Released HSP are engulfed and processed by immature dendritic cells (iDC) attracted to the tissue damage. The inflamma- tory microenvironment created by the BAG and the sub- sequent allo-rejection response amplified by the priming induces DC1 maturation. These DC1 display processed tumor antigens on upregulated MHCI/II and express co-stimulatory CD80/86 enabling priming of a tu- mor-specific Th1/CTL response. Intravenous BAG express- ing CD40L and IFN-ϒ activates allo-specific and tu- mor-specific memory cells, permitting trafficking to tumor sites. The host rejection of BAG releases endoge- nous danger signals, creating a sustained systemic in- flammatory cytokine release which serves to counter-reg- ulate immunosuppressive mechanisms. Longitudinal CT scans biopsies, PBMC and serum samples are collected for analysis to verify immune events within each phase of the protocol. Clinical trial information: NCT0238044 Th1 CD8 CD4 CTL Th1 CTL Th1 CTL CTL Th1 Th1 CTL The objective of Part A of the study is to test various dose/dose-frequency cohorts to determine if any provide anti-tumor effects (pathologically, radiologically or biologically) and subsequently select from the cohorts that provide anti-tumor effects, the cohort with the best tolerability to test in an additional 25 subjects in Part B. The cohorts under investigation are shown below. Dosing Schedule Introduction This trial-in-progress is evaluating a new therapeutic cancer vaccine platform. The clinical benefit of thera- peutic cancer vaccines has been almost negligible de- spite the use of a variety of innovative technologies, an- tigens, adjuvants and delivery methods. Therapeutic vaccine efficacy is hindered by several factors, including: (1) tumor immunoavoidance through tumor-induced impairment of antigen presentation, downregulation of MHC molecules, defective co-stimulation, expres- sion of checkpoint molecules and tumor production of immunosuppressive cytokines (such as IL-10 and TGF-β); (2) poor tumor “self” antigen immunogenicity; (3) outgrowth of immune-resistant tumor clones due to immunoediting; (4) tumor-influenced activation of tolerance-induc- ing immunosuppressive circuits, including induction of CD4+, CD25+, FoxP3+ regulatory T cells (Treg), sup- pressive natural killer T cells (NKT2), myeloid-derived suppressor cells (MDSC), tumor associated M2 macro- phages (TAM), Th2/Tr1 T-cells, and immunosuppres- sive subsets of mature dendritic cells (DC2); (5) tumor-induced immune-deviation to Type 2-dom- inated immunity both systemically and in the tumor microenvironment; and (6) immunosuppression due to heavy medical pre- treatment and the presence of large tumor burdens. Rather than attempt to improve vaccine platforms that had previously failed, we aimed to develop a new therapeutic vaccine platform. We based our vaccine platform design upon the already proven anti-tumor immune cascades that occur after allogeneic, non-my- eloablative, stem cell transplant (ASCT) procedures. After ASCT, a graft vs. tumor (GVT) cascade occurs which has proven ability to kill refractory hematologi- cal malignancies and chemotherapy-resistant meta- static solid tumors, including metastatic CRC. Howev- er, this beneficial anti-tumor effect is linked to graft vs. host disease (GVHD). The high morbidity and mortali- ty of GVHD severely limits the use of ASCT procedures for treating solid tumors. Despite decades of research, the separation of the beneficial GVT effects from the devastating effects of GVHD remains elusive. We developed a vaccine platform that provides the GVT effects of ASCT without GVHD, chemotherapy conditioning or need for tissue-matched donors. This is accomplished by reversing the immunological flow of the GVT/GVHD effects. Instead of the flow emanat- ing from engrafted donor cells, the flow instead was engineered to emanate from an intact host immune system. After introduction of an allograft, the intact host immune system responds with a non-toxic host vs. graft (HVG) rejection which can serve as an adju- vant for eliciting a linked host vs. tumor (HVT) effect. The chemotherapy conditioning regime and GVHD are essential for eliciting GVT effects after ASCT. In order to elicit host-mediated HVT effects upon HVG rejection without prior engraftment, the allograft had to be bioengineered to replace the contributions of chemotherapy conditioning and GVHD. Chemothera- py conditioning and GVHD cause tissue damage which results in: release of endogenous danger asso- ciated molecular patterns (DAMP); translocation of LPS from the GI tract; and, a Type 1 “cytokine storm” with IFN-γ as the key component. These factors pro- mote development of cellular immunity and down-regulate immunosuppression and immu- noavoidance mechanisms. BAG are bioengineered to the provide these same es- sential components. BAG cells are intentionally mis- matched, ex-vivo differentiated, activated memory Th1 cells which express Type 1 cytokines such as IFN-γ and express high density CD40L. Upon rejection, BAG release endogenous DAMP. CD40L has the same immune effects as LPS. AUTHORS 1 Banner MD Anderson Cancer Center, Gilbert, AZ 2 Immunovative Clinical Research, Inc., Mesa, AZ 3 Immunovative Therapies, Ltd, Jerusalem, Israel 4 [email protected] 5 [email protected] Madappa N. Kundranda 1,4 , Tomislav Dragovich 1 , Andrew Price 1 , Philippe Lanauze 1 , Miriam Bloch 2 , Michael Bishop 2 , John Chang 1 and Michael Har-Noy 3,5 Abstract # 202348 Tumor-Specific Immunity The titers of activated, tumor-specific non-memory and memory Th1/CTL cells is increased. CTL Th1 CTL Th1 Allo-specific Th1 and CTL T-cells traffic to bloodstream Tumor-specific Th1 and CTL T-cells traffic to bloodstream Tumor-specific Th1 and CTL T-cells traffic to bloodstream Allo-specific and Tumor-specific Th1 and CTL T-cells traffic to bloodstream Methods BAG are produced in Israel under Good Manufactur- ing Practices (GMP). Each lot is quality checked to assure it meets pre-determined identity, function and safety criteria. BAG cells are manufactured from blood collected from healthy donors that have been screened and tested pursuant to 21 CFR 1271. Puri- fied CD4+ cells are expanded and differentiated ex-vi- vo in a bioreactor. After 9 days, an intermediate cell is produced, which is aliquoted into single dose vials Trial Design Third-line metastatic colorectal cancer subjects that have progressed after an oxaliplatin- and irinotec- an-based chemotherapy are eligible. Minimal expo- sure to regorafenib or TAS 102 is allowed. The objec- tive of Part A of the study is to test various dose/dose-frequency cohorts (see Figure below) in a 3+3 design to determine the dosing pattern that has the best response/toxicity ratio. In Part B of the study, one cohort will be evaluated in 25 additional subjects. Dose Limiting Toxicity (DLT) is evaluated prior to cohort advance. DLT is defined as the occurrence of any of the following within 48h of dosing: Any ≥ Grade 3 local injection site reaction; Any ≥ Grade 3 infusion related symptoms of dura- tion > 2 days; Any ≥ Grade 4 infusion reaction of any duration; Or any of the following up to 28 days following the last dosing: Any ≥ Grade 3 graft vs. host disease; Any toxicity ≥ Grade 3 that is probably or definitely related to the study drug; Any toxicity not present at baseline that occurs at ≥ Grade 4; Any autoimmune toxicity ≥ Grade 3 not present at baseline. and stored frozen. When required in the clinic, the frozen vials are thawed and the intermediated cells incubated with CD3/CD28 microbeads (1:1 ratio). After 4h, the intermediate cells differentiate into BAG. The BAG cells with microbeads attached are formulat- ed in PlasmaLyteA with 1% HSA and loaded at 1 x 107 cells/ml into syringes. The loaded syringes are shipped refrigerated to clinical sites pursuant to the dosing schedule and are stable for 120h at 20-80C. Allo Antigen Allo Antigen Tumor Antigen Tumor Antigen Type 1 Cytokines Tumor Cohort Day 0 Day 3 Day 7 Day 10 Day 14 Day 17 Day 21 Day 28 Day 49 Day 56 Day 77 Day 84 Day 112 A — Control schedule, previously tested ID -1 ml ID- 1 ml ID - 1 ml Cryo + IT - 3 ml IV - 5 ml IV - 5 ml IV - 5 ml A-1 — Low dose treatment w/o cryoablation ID - 0.5 ml ID - 0.5 ml ID - 0.5 ml ID - 0.5 ml IV - 3 ml IV - 3 ml IV - 3 ml IV - 3 ml A-2 — Low dose treatement w/ cryoablation ID - 0.5 ml ID - 0.5 ml ID - 0.5 ml Cryo + IT - 1 ml IV - 3 ml IV - 3 ml IV - 3 ml IV - 3 ml A-3 — Increased ID and IV dosing w/o cryoablation ID - 1 ml ID - 1 ml ID - 1 ml ID - 1 ml IV - 5 ml IV - 5 ml IV - 5 ml IV - 5 ml A-4 — Increased ID and IV dosing w/ cryoablation ID - 1 ml ID - 1 ml ID - 1 ml Cryo + IT - 3 ml IV - 5 ml IV - 5 ml IV - 5 ml IV - 5 ml B-ID — Frequency escalation ID - 1 ml ID - 1 ml ID - 1 ml ID - 1 ml Cryo + IT - 3 ml IV - 5 ml IV - 5 ml IV - 5 ml C-IT Frequency escalation ID - 1 ml ID - 1 ml ID - 1 ml ID - 1 ml Cryo + IT - 3 ml IT - 3 ml IV - 5 ml IV - 5 ml IV - 5 ml D-IV — Frequency escalation during priming ID - 1 ml ID - 1 ml ID - 1 ml + IV - 3 ml ID - 1 ml + IV - 3 ml Cryo + IT - 3 ml IV - 5 ml IV - 5 ml IV - 5 ml E-IV Frequency escalation during priming and vaccination ID - 1 ml ID - 1 ml ID - 1 ml + IV - 3 ml ID - 1 ml + IV - 3 ml Cryo + IT - 3 ml + IV - 3 ml IV - 5 ml IV - 5 ml IV - 5 ml F-IT and IV — Frequency escalation ID - 1 ml ID - 1 ml ID - 1 ml + IV - 3 ml ID - 1 ml + IV - 3 ml Cryo+ IT - 3 ml + IV - 3 ml IV - 3 ml IV - 5 ml IV - 5 ml IV - 5 ml Notes: | For Cohorts B-F, ID doses on days 0 and 3 are given in the same location and ID doses on days 7 and 10 are given in the same location | For Cohort C, IT injections are given into the same ablated lesion on days 14 and 17

Upload: others

Post on 25-Jul-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Phase I/IIb Trial: In-Situ Vaccine for Metastatic Colorectal Cancer · 2018-01-21 · CD8 As iDC engulfs HSP, it matures to DC1 Dendritic Cell Maturation: The HSP are engulfed by

CD8

As iDC engulfs HSP, it matures to DC1

Dendritic Cell Maturation:The HSP are engulfed by iDC which process the alloantigens chaperoned on the released HSP. In the presence of DAMP, the iDC mature to IL-12+ DC1. These activated DC1 tra�c to the draining lymph nodes, upregulate CD80/86 co-stimulatory molecules and present the processed alloantigens on MHCI and MHCII.

Initiation:In the priming phase, there are multiple intradermal (ID) injections of BAG cells. GM-CSF produced by the BAG, attracts NK cells and immature DC (iDC), such as Langerhans’s cells, to the injection site. NK cells in the presence of IFN-γ upregulate NKG2D-L expression enabling recognition and lysis of the allogeneic NKG2D+ BAG cells. Lysing of the BAG cells causes release of endogenous DAMP and HSP chaperones of the alloantigens into the microenvironment, creating an in-situ anti-alloantigen vaccine. iDC engulf and process the alloantigens to initiate the anti-alloantigen immune cascade. Once allo-speci�c immunity is established, subsequent ID BAG injections can also be rejected by the allo-speci�c Th1/CTL.

Non-Speci�c Lysis of Tumor CellsThe extravasating waves of activated NK cells and activated Type 1 memory cells encounter a tumor microenvironment that is profoundly immunosuppressive, with resident myeloid-derived suppressor cells (MDSC), Treg, Th2/Tr1, tumor-associated macrophages (TAM) and cancer-associated �broblasts (CAF). These suppressor cells maintain a Type 2 cytokine environment dominated by IL-10, TGF-β and IL-6 which suppresses cellular immune function. In a Type 2 environment, tumor cells down-regulate MHC I and co-stimulatory molecule expression and upregulate expression of checkpoint molecules (e.g., CTLA4/PD-L1) all which inhibit recognition and attack by CTL. In�ltrating activated NK cells and Type 1 memory cells condition the tumor microenvironment by contributing Type 1 cytokines such as IFN-γ , TNF-α and IL-2. These Type 1 cytokines cause di�erentiation of resident macrophages to the M1 tumoricidal phenotype, counter-regulate the e�ects of in�ltrating suppressor cells, and upregulate counter death receptors, MHC I and co-stimulatory molecules on tumor cells. Under these conditions, non-speci�c tumor lysis can occur through activated NK cells, M1 macrophages and activated memory cells. Tumor lysis releases endogenous DAMP and endogenous tumor neoantigens chaperoned on HSP. The released HSP are engulfed and processed by iDC, which in the presence of DAMP and Type 1 cytokines mature to IL-12+ DC1 and tra�c to the draining lymph node to initiate the tumor-speci�c immune cascade.

Tumor

Bloodstream ActivationOnce in circulation, the activated, non-memory, allo-speci�c Th1 and CTL T-cells produce IFN-γ and express CD40L. This causes activation of circulating NK cells and memory T-cells. Activated NK cells and memory cells tra�c to the tumor sites. Activated memory cells express ‘death receptors’ such as FasL, TRAIL and TWEEK which can non-speci�cally kill tumors in a Type 1 cytokine environment. Subsequent BAG injections release additional waves of activated, non-memory, allo-speci�c Th1 and CTL T-cells into circulation which, in turn, causes new waves of activated NK cells and memory cells to in�ltrate tumor sites. After each BAG injection, a portion of the activated, non-memory, allo-speci�c Th1 and CTL will di�erentiate into memory cells. As the titer of circulating memory allo-speci�c Th1 and CTL increases in circulation, each wave of activated, non-memory, allo-speci�c Th1 and CTL will non-speci�cally activate increasing numbers of allo-speci�c Th1 and CTL cells from the circulating memory pool . As increased numbers of activated NK and Th1/CTL memory cells enter the tumor microenvironment, the production of Type 1 cytokines modulates the tumor environment from Type 2 to Type 1 dominance.

Priming PhaseIn the priming phase, BAG cells are injected intradermally in order to elicit high titers of allo-speci�c Th1/CTL in circulation. These activated T-cells in turn non-speci�cally activate circulating NK cells, M1 macrophages and memory T-cells through production of IFN-γ and CD40:CD40L interaction. All these non-speci�c e�ector cells tra�c to tumor sites and condition the local microenvironment with Type 1 cytokines In the presence of Type 1 cy-tokines, all these non-speci�cally activated cells can lyse tumor cells. This tumor lysis results in the release of endogenous heat shock proteins (HSP) and danger-associated molecular patterns (DAMP) from the lysed tumors. HSP chaperone the complete tumor antigen repertoire, including neoantigens. Therefore, released HSP in the context of the Type 1 cytokine environment and DAMP, provides an in-situ vaccine with tumor neoantigen and ad-juvant danger signals for eliciting tumor-speci�c immunity.

Vaccination PhaseThe vaccination phase serves to increase the in�ltration and e�ector function of tumor antigen speci�c Th1/CTL within the tumor lesions and development of tumor-speci�c memory. Each priming injection of BAG cells causes waves of activated non-memory allospeci�c Th1/CTL and memory allospeci�c Th1/CTL. Each wave of non-memory allospeci�c Th1/CTL causes activation and extravasation of memory allospeci�c Th1/CTL and NK cells which conditions the tumor mi-croenvironment with Type 1 cytokines. In this environment, NK cells and activated memory allo-speci�c Th1/CTL along with resident M1 macrophages mediate non-speci�c tumor lysis. The non-speci�c lysis of tumor cells releases endogenous chaperoned tumor neoantigens and DAMP. The presence of tumor neoantigens and DAMP in a Type 1 environment creates an in-situ anti-tumor vaccine. iDC process the tumor antigens, tra�c to lymph nodes and cause release of activated non-memory tumor-speci�c Th1/CTL in circulation. These cells activate circulating memory cells the same as activated non-memory allo-speci�c cells. As additional waves of tumor lysis occur, a portion of the activated non-memory tumor-speci�c Th1/CTL di�erentiate to memory cells. As the titers of both allo-speci�c and tumor-speci�c memory cells increase in cir-culation, each wave of activated non-memory allo-speci�c and tumor speci�c cells activates increased numbers of allo-speci�c and tumor-speci�c memory cells.

DC1 migrates to draining lymph node

DC1 migrates to draining lymph node

Alloantigen Chaperoned on HSP Tumor antigen Chaperoned on HSP

iDC

Allo-Speci�c CTL and TH1 T-Cell Activation:In the draining lymph nodes, the DC1 interact with naïve CD4+ and CD8+ T-cells and produce IL-12. In the presence of IL-12, naïve CD4+ and CD8+ T-cells, which recognize the alloantigens, di�erentiate to allo-speci�c Th1 and CTL, respectively. These activated, allo-speci�c T-cells produce IFN-γ , upregulate CD40L and down-regulate CD62L. Down-regulation of CD62L permits their entry into the circulation.

DC1

MHC I molecule

MHC II moleculeCo-stimulatory

molecule CD 80/86

Education signals are received as naïve T-Cells

interact with their cognate antigens on the MHC I and

MHC II molecules

Arming signals are received via CD80/86 molecules on

the DC1 to CD28 surface molecules on the

naïve T-Cells

Education signals are received as naïve T-Cells

interact with their cognate antigens on the MHC I and

MHC II molecules

Arming signals are received via CD80/86 molecules on

the DC1 to CD28 surface molecules on the

naïve T-Cells

Allo-Speci�c Immunity

Allo-Speci�c Immunity

Tumor-Speci�c Immunity

Tumor-Speci�c Immunity

Tumor-Speci�c Immunity

Innate Immunity

LYMPH NODE

DC1

LYMPH NODE

DC1

Adaptive Immunity

Adaptive Immunity

iDC

As iDC engulfs HSP, it matures to DC1

iDC engulfs HSP and matures to DC1

iDC engulfs HSP and matures to DC1

iDC engulfs HSP and matures to DC1

DC1

MHC I

MHC II

Ampli�cationThe in-situ vaccine e�ect caused by the non-speci�c lysis of tumor cells in a Type 1 cytokine envi-ronment, produces new waves of activated non-memory tumor-speci�c Th1/CTL. As this cas-cade feeds forward, a proportion of the non-memory tumor-speci�c Th1/CTL di�erentiate to memory cells. Each new wave of activated non-memory allo-speci�c and/or non-memory tu-mor-speci�c Th1/CTL activates these circulating memory cells causing them to tra�c to tumor lesions. To amplify these e�ects, a select tumor lesion is killed through use of a percutaneous cryoablation followed immediately by injection of BAG cells. The cryoablation freeze-thaw cycles cause massive release of endogenous DAMP and HSP chaperones from lysed tumor cells. The insertion of the allogeneic BAG cells into the lesion recruits allo-speci�c Th1/CTL memory cells to reject the BAG, releasing additional DAMP and Type 1 cytokines. The immune stimulato-ry e�ects of the alloantigens mixed with the tumor antigens in the context of Type 1 cytokines and DAMP, ampli�es the development of tumor-speci�c memory Th1/CTL.

Activation and BoosterThe priming and vaccination phases serve to increase the titers of allo-speci�c and tumor-specif-ic Th1/CTL memory cells. BAG produce IFN-ϒ and express high density CD40L. Intravenous infu-sion of BAG increases the extravasation of these memory cells to tumor lesions through CD40L:CD40 interaction in a Type 1 cytokine environment. The allo-speci�c rejection response to BAG in circulation serves to release endogenous DAMP which activates circulating monocytes causing a Type 1 “cytokine storm”. The Type 1 cytokines serve to counter-regulate immunosup-pressive and immunoavoidance mechanisms and support on-going anti-tumor cellular immune responses. In the event that the Type 1 cytokine storm wanes between dosing and prior to com-plete elimination of tumor, the anti-tumor e�ects of can be re-activated through subsequent BAG intravenous booster infusions.

In the event of over-activation of the immune system, resulting in immune-mediated toxicity, steroids can be administered. Since dexamethasone causes apoptosis of activated Th1/CTL, it may serve as a “reversal” agent.

CD80/86

DC1 migrates to draining lymph node

DC1 migrates to draining lymph node

Adaptive Immunity

iDC DC1

iDC DC1

Adaptive Immunity

DC1 migrates to draining lymph node

iDCDC1

Bioengineered Allogeneic Graft (BAG) Cells:Intentionally-mismatched, ex-vivo expanded and di�er-entiated memory CD4+ Th1 cells with anti-CD3/CD28 microbeads attached that express high density CD40L, MHC II, NKG2D and Type 1 cytokines including IFN-γ, TNF-α and GM-CSF.

Percutaneous Cryoablation

Tumor

Allogeneic BAG Cell Injection into Lesion

Adaptive Immunity

Counter-regulate Immunosuppressive and Immunoavoidance Th2 Mechanisms

Adaptive Immunity

NK

NK

CD4

DiscussionThis novel therapeutic vaccine platform is being tested for ability to elicit immune-mediated anti-tu-mor e�ects in third-line MSI-Low metastatic CRC pa-tients not responsive to checkpoint blockade immu-notherapy. The mechanism of the vaccine acts to pro-vide feed-forward amplifying waves of innate anti-tu-mor e�ects which transition to waves of tumor-specif-ic immune e�ects. The mechanism also incorporates a strategy for counter-regulating the immunosuppres-sive tumor microenvironment. Some cohorts incorpo-rate an in-situ vaccination strategy that combines per-cutaneous cryoablation of a tumor lesion followed by intratumoral injection of BAG cells. In-situ vaccination provides additional ampli�cation of the anti-tumor e�ects of the vaccine. In order to evaluate anti-tumor responses and distinguish pseudoprogression from actual progression, the protocol evaluates longitudi-nal simultaneous CT scans and biopsy samples of a select tumor lesion. Peripheral blood samples are also collected for evaluation of various biomarkers and immune parameters.

Phase I/IIb Trial: In-Situ Vaccine for Metastatic Colorectal Cancer

AbstractBackground: Except for MSI-H tumors, colorectal cancer (CRC) does not respond to immunotherapy. CRC does respond to the graft vs. tumor (GVT) immune e�ect that occurs after allo-geneic stem cell transplantation (ASCT). GVT is associated with graft vs. host disease (GVHD) toxicity which limits the clinical application. A bioengineered allograft (BAG) has been developed which can elicit host-mediated GVT-like e�ects without GVHD toxicity, chemotherapy condition-ing or a HLA-matched donor. BAG are Th1 memory cells derived from blood of healthy donors with CD3/CD28 mi-crobeads attached. These cells have immunomodulatory properties which enable modulation of Type 1/Type 2 bal-ance and dysregulation of immunosuppressive circuits. We are evaluating the safety and e�cacy of BAG in third-line metastatic CRC.

Methods: The study uses a standard 3+3 design followed by an ex-pansion phase with the optimal dosing pattern. The pro-tocol has four components: (A) priming; (B) in-situ vacci-nation; (C) activation; and (D) booster. Priming involves intradermal injections of BAG cells which activates NK cells and develops allo-speci�c Th1/CTL immunity. In-situ vaccination involves tumor cryoablation to release en-dogenous heat shock proteins (HSP) which chaperone tu-mor-speci�c neoantigens, followed immediately by the intralesional injection of BAG cells as adjuvant. Released HSP are engulfed and processed by immature dendritic cells (iDC) attracted to the tissue damage. The in�amma-tory microenvironment created by the BAG and the sub-sequent allo-rejection response ampli�ed by the priming induces DC1 maturation. These DC1 display processed tumor antigens on upregulated MHCI/II and express co-stimulatory CD80/86 enabling priming of a tu-mor-speci�c Th1/CTL response. Intravenous BAG express-ing CD40L and IFN-ϒ activates allo-speci�c and tu-mor-speci�c memory cells, permitting tra�cking to tumor sites. The host rejection of BAG releases endoge-nous danger signals, creating a sustained systemic in-�ammatory cytokine release which serves to counter-reg-ulate immunosuppressive mechanisms. Longitudinal CT scans biopsies, PBMC and serum samples are collected for analysis to verify immune events within each phase of the protocol.

Clinical trial information: NCT0238044

Th1

CD8

CD4 CTLTh1

CTLTh1

CTL

CTLTh1Th1 CTL

The objective of Part A of the study is to test various dose/dose-frequency cohorts to determine if any provide anti-tumor e�ects (pathologically, radiologically or biologically) and subsequently select from the cohorts that provide anti-tumor e�ects, the cohort with the best tolerability to test in an additional 25 subjects in Part B. The cohorts under investigation are shown below.

Dosing Schedule

IntroductionThis trial-in-progress is evaluating a new therapeutic cancer vaccine platform. The clinical bene�t of thera-peutic cancer vaccines has been almost negligible de-spite the use of a variety of innovative technologies, an-tigens, adjuvants and delivery methods. Therapeutic vaccine e�cacy is hindered by several factors, including:

(1) tumor immunoavoidance through tumor-induced impairment of antigen presentation, downregulation of MHC molecules, defective co-stimulation, expres-sion of checkpoint molecules and tumor production of immunosuppressive cytokines (such as IL-10 and TGF-β);

(2) poor tumor “self” antigen immunogenicity;

(3) outgrowth of immune-resistant tumor clones due to immunoediting;

(4) tumor-in�uenced activation of tolerance-induc-ing immunosuppressive circuits, including induction of CD4+, CD25+, FoxP3+ regulatory T cells (Treg), sup-pressive natural killer T cells (NKT2), myeloid-derived suppressor cells (MDSC), tumor associated M2 macro-phages (TAM), Th2/Tr1 T-cells, and immunosuppres-sive subsets of mature dendritic cells (DC2);

(5) tumor-induced immune-deviation to Type 2-dom-inated immunity both systemically and in the tumor microenvironment; and

(6) immunosuppression due to heavy medical pre-treatment and the presence of large tumor burdens.

Rather than attempt to improve vaccine platforms that had previously failed, we aimed to develop a new therapeutic vaccine platform. We based our vaccine platform design upon the already proven anti-tumor immune cascades that occur after allogeneic, non-my-eloablative, stem cell transplant (ASCT) procedures. After ASCT, a graft vs. tumor (GVT) cascade occurs which has proven ability to kill refractory hematologi-cal malignancies and chemotherapy-resistant meta-

static solid tumors, including metastatic CRC. Howev-er, this bene�cial anti-tumor e�ect is linked to graft vs. host disease (GVHD). The high morbidity and mortali-ty of GVHD severely limits the use of ASCT procedures for treating solid tumors. Despite decades of research, the separation of the bene�cial GVT e�ects from the devastating e�ects of GVHD remains elusive.

We developed a vaccine platform that provides the GVT e�ects of ASCT without GVHD, chemotherapy conditioning or need for tissue-matched donors. This is accomplished by reversing the immunological �ow of the GVT/GVHD e�ects. Instead of the �ow emanat-ing from engrafted donor cells, the �ow instead was engineered to emanate from an intact host immune system. After introduction of an allograft, the intact host immune system responds with a non-toxic host vs. graft (HVG) rejection which can serve as an adju-vant for eliciting a linked host vs. tumor (HVT) e�ect.

The chemotherapy conditioning regime and GVHD are essential for eliciting GVT e�ects after ASCT. In order to elicit host-mediated HVT e�ects upon HVG rejection without prior engraftment, the allograft had to be bioengineered to replace the contributions of chemotherapy conditioning and GVHD. Chemothera-py conditioning and GVHD cause tissue damage which results in: release of endogenous danger asso-ciated molecular patterns (DAMP); translocation of LPS from the GI tract; and, a Type 1 “cytokine storm” with IFN-γ as the key component. These factors pro-mote development of cellular immunity and down-regulate immunosuppression and immu-noavoidance mechanisms.

BAG are bioengineered to the provide these same es-sential components. BAG cells are intentionally mis-matched, ex-vivo di�erentiated, activated memory Th1 cells which express Type 1 cytokines such as IFN-γ and express high density CD40L. Upon rejection, BAG release endogenous DAMP. CD40L has the same immune e�ects as LPS.

A U T H O R S

1Banner MD Anderson Cancer Center, Gilbert, AZ

2Immunovative Clinical Research, Inc., Mesa, AZ

3Immunovative Therapies, Ltd, Jerusalem, Israel

[email protected]

5 [email protected]

Madappa N. Kundranda1,4, Tomislav Dragovich1, Andrew Price1, Philippe Lanauze1, Miriam Bloch2,

Michael Bishop2, John Chang1 and Michael Har-Noy3,5

Abstract # 202348

Tumor-Speci�c Immunity

The titers of activated, tumor-speci�c non-memory and

memory Th1/CTL cells is increased.

CTLTh1

CTLTh1

Allo-speci�c Th1 and CTL T-cells tra�c to bloodstream

Tumor-speci�c Th1 and CTL T-cells tra�c to bloodstream

Tumor-speci�c Th1 and CTL T-cells tra�c to bloodstream

Allo-speci�c and Tumor-speci�c Th1 and CTL T-cells tra�c to bloodstream

MethodsBAG are produced in Israel under Good Manufactur-ing Practices (GMP). Each lot is quality checked to assure it meets pre-determined identity, function and safety criteria. BAG cells are manufactured from blood collected from healthy donors that have been screened and tested pursuant to 21 CFR 1271. Puri-�ed CD4+ cells are expanded and di�erentiated ex-vi-vo in a bioreactor. After 9 days, an intermediate cell is produced, which is aliquoted into single dose vials

Trial DesignThird-line metastatic colorectal cancer subjects that have progressed after an oxaliplatin- and irinotec-an-based chemotherapy are eligible. Minimal expo-sure to regorafenib or TAS 102 is allowed. The objec-tive of Part A of the study is to test various dose/dose-frequency cohorts (see Figure below) in a 3+3 design to determine the dosing pattern that has the best response/toxicity ratio. In Part B of the study, one cohort will be evaluated in 25 additional subjects.

Dose Limiting Toxicity (DLT) is evaluated prior to cohort advance. DLT is de�ned as the occurrence of any of the following within 48h of dosing:

• Any ≥ Grade 3 local injection site reaction; • Any ≥ Grade 3 infusion related symptoms of dura-

tion > 2 days;• Any ≥ Grade 4 infusion reaction of any duration;

Or any of the following up to 28 days following the last dosing:

• Any ≥ Grade 3 graft vs. host disease;• Any toxicity ≥ Grade 3 that is probably or de�nitely

related to the study drug;• Any toxicity not present at baseline that occurs at ≥

Grade 4;• Any autoimmune toxicity ≥ Grade 3 not present at

baseline.

and stored frozen. When required in the clinic, the frozen vials are thawed and the intermediated cells incubated with CD3/CD28 microbeads (1:1 ratio). After 4h, the intermediate cells di�erentiate into BAG. The BAG cells with microbeads attached are formulat-ed in PlasmaLyteA with 1% HSA and loaded at 1 x 107 cells/ml into syringes. The loaded syringes are shipped refrigerated to clinical sites pursuant to the dosing schedule and are stable for 120h at 20-80C.

Allo Antigen

Allo AntigenTumor

Antigen

Tumor Antigen

Type 1 Cytokines

Tumor

Cohort Day 0 Day 3 Day 7 Day 10 Day 14 Day 17 Day 21 Day 28 Day 49 Day 56 Day 77 Day 84 Day 112

A — Control schedule, previously tested ID -1 ml ID- 1 ml ID - 1 ml Cryo + IT - 3 ml IV - 5 ml IV - 5 ml IV - 5 ml

A-1 — Low dose treatment w/o cryoablation ID - 0.5 ml ID - 0.5 ml ID - 0.5 ml ID - 0.5 ml IV - 3 ml IV - 3 ml IV - 3 ml IV - 3 ml

A-2 — Low dose treatement w/ cryoablation ID - 0.5 ml ID - 0.5 ml ID - 0.5 ml Cryo + IT - 1 ml IV - 3 ml IV - 3 ml IV - 3 ml IV - 3 ml

A-3 — Increased ID and IV dosing w/o cryoablation ID - 1 ml ID - 1 ml ID - 1 ml ID - 1 ml IV - 5 ml IV - 5 ml IV - 5 ml IV - 5 ml

A-4 — Increased ID and IV dosing w/ cryoablation ID - 1 ml ID - 1 ml ID - 1 ml Cryo + IT - 3 ml IV - 5 ml IV - 5 ml IV - 5 ml IV - 5 ml

B-ID — Frequency escalation ID - 1 ml ID - 1 ml ID - 1 ml ID - 1 ml Cryo + IT - 3 ml IV - 5 ml IV - 5 ml IV - 5 ml

C-IT — Frequency escalation ID - 1 ml ID - 1 ml ID - 1 ml ID - 1 ml Cryo + IT - 3 ml IT - 3 ml IV - 5 ml IV - 5 ml IV - 5 ml

D-IV — Frequency escalation during priming ID - 1 ml ID - 1 ml ID - 1 ml + IV - 3 ml ID - 1 ml + IV - 3 ml Cryo + IT - 3 ml IV - 5 ml IV - 5 ml IV - 5 ml

E-IV — Frequency escalation during priming and vaccination ID - 1 ml ID - 1 ml ID - 1 ml + IV - 3 ml ID - 1 ml + IV - 3 ml Cryo + IT - 3 ml + IV - 3 ml IV - 5 ml IV - 5 ml IV - 5 ml

F-IT and IV — Frequency escalation ID - 1 ml ID - 1 ml ID - 1 ml + IV - 3 ml ID - 1 ml + IV - 3 ml Cryo+ IT - 3 ml + IV - 3 ml IV - 3 ml IV - 5 ml IV - 5 ml IV - 5 ml

Notes: | For Cohorts B-F, ID doses on days 0 and 3 are given in the same location and ID doses on days 7 and 10 are given in the same location | For Cohort C, IT injections are given into the same ablated lesion on days 14 and 17