phase ii trial of a personalized˜vaccine for advanced hcc€¦ · this trial-in-progress is...

1
hsp70, hsp90, gr94/gp96 and calreticulin. This Chaperone-Rich Cell Lysate (CRCL) is quality tested to assure the identity, purity, sterility, low endo- toxin and integrity of the chaperoned tumor anti- gens. The purified HSP are injected intradermally together with BAG cells. In order to customize an immune response against a patient’s own tumor cells and increase the titers of activated allo-specific and tumor-specific Th1/CTL in circulation, a biopsy sample of a tumor is collect- ed and sent to Jerusalem for processing. In Jerusalem, the tumors are processed under Good Manufacturing Practices (GMP) to isolate endogenous HSP, including Phase II Trial of a Personalized Vaccine for Advanced HCC Abstract Background: The chronic inflammation and viral infection associated with HCC combined with liver tolerogenic mechanisms creates a profoundly immunosuppressive microenviron- ment. Modulating the microenvironment with check- point blockade benefits ~20% of patients. The non-re- sponders and those with poor liver function represent an unmet medical need. Checkpoint blockade requires ef- fector cells to be resident within tumors. However, the majority of HCC lesions lack infiltrating effector cells. Therefore, a strategy for amplifying the tumor-specific immune response, while counteracting the immunosup- pressive mechanisms may provide an improved immuno- therapy. Endogenous heat shock proteins (HSP) chaper- one tumor neoantigens. A process enabling the concen- tration of HSP such as calreticulin, hsp70, hsp90 and gr94/gp96 (CRCL) from tumor biopsy samples has been developed and serves as a source of tumor neoantigen for vaccination. In addition, a bioengineered allogeneic allograft (BAG) derived from healthy blood donors with potent immunomodulatory and counter-regulatory properties has been developed as an adjuvant. The com- bination of CRCL with BAG is being evaluated as a strate- gy to amplify tumor-specific immunity and counter-regu- late the immunosuppressive microenvironment in ad- vanced HCC. Methods: An open-label Phase II clinical trial evaluating the safety and efficacy of CRCL+BAG vaccine in Child-Pugh A/B ad- vanced or metastatic HCC w/ or w/o Sorafenib is being conducted at Khon Kaen University Medical Center in Thailand . Approximately 6-10 biopsy cores are collected at baseline. The tumor sampe is lysed ex-vivo and the en- dogenous CRCL purified using an isoelectric focusing technique. Subjects are first primed with multiple intra- dermal injections of BAG cells alone to increase the titer of allo-specific Th1/CTL cells. Next, multiple intradermal injections of BAG+CRCL are administered to elicit in- creased titers of tumor-specific Th1/CTL cells. Subsequent intravenous infusions of BAG cells activate allo-specific and tumor-specific memory cells, enabling their infiltra- tion into tumor lesions. The BAG allo-rejection response creates a type I ‘cytokine storm’ which serves to down-reg- ulate suppressor circuits. Response is evaluated by mRE- CIST. Longitudinal CT scans with concurrent biopsies as well as multiple collections of PBMC and plasma permits immunomonitoring of these immune mechanisms. Clinical trial information: NCT02409524 Discussion Current therapies for advanced HCC are marginally effective and exacerbate underlying liver disease. The inherent immune tolerance of the liver suppresses cellular immunity and permits HCC progression and spread. The liver is exposed to abundant xenoantigens from the gut via the portal vein. These xenoantigens tend to polarize the liver microenvironment to Type 2 which induces immune tolerance and recruitment of suppressor cells, such as Treg and MDSC. In addition, the Type 2 conditions upregulate expression of check- point molecules, such programmed death ligand 1 (PD-L1), which contributes to further weaken anti-tu- mor immune responses. Nonparenchymal cells, including stellate cells, hepatic dendritic cells and liver sinusoidal endothelial cells also can induce tolerance and/or apoptosis of effector Th1/CTL cells, further exacerbating the hostile envi- ronment for cellular anti-tumor immune function. Dosing Schedule Introduction This trial-in-progress is evaluating a new therapeutic cancer vaccine platform in advanced hepatocellular carcinoma (HCC). HCC is one of the most common ma- lignancies worldwide and is endemic in Southeast Asia due to the high incidence of hepatitis B and hep- atitis C. Some of the highest incidence rates occur within the Khon Kaen region of Thailand, which is the site of this study. In Thailand, most HCC patients pres- ent to the clinic with advanced Barcelona Clinic Liver Cancer [BCLC] stage C disease. These patients have limited therapeutic options. Approved therapies including sorafenib, regorafenib and nivolomab have response rates <20% and only a modest benefit in terms of overall survival. In addition, these treatments are generally too expensive for widescale use in the Thai population. Since HCC has been shown to be immunogenic and is responsive to checkpoint blockade, therapeutic vacci- nation represents a promising treatment option for these patients. Successful development of a thera- peutic vaccine has potential to increase survival and to have lower cost than current options. However, while past therapeutic vaccine immunotherapy strat- egies against HCC have demonstrated tumor-specific T cell responses, these responses were often not robust enough to induce clinical responses. 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-inducing 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-domi- nated immunity both systemically and in the tumor microenvironment; and (6) immunosuppression due to the immune privi- leged nature of the liver biology. In chronically inflamed livers, genetic and epigenetic changes underlie oncogenic transformation and pro- duce mutations which can be recognized as neoanti- gens by the immune system. The complete antigenic repertoire of tumor cells is associated with endoge- nous heat shock proteins (HSP). We developed a method of purifying HSP from tumor biopsy samples resulting in a vaccine preparation enriched in hsp70, hsp90, gr94/gp96 and calreticulin (known as chaper- one rich cell lysate or “CRCL”). CRCL provides a source of patient-specific tumor neoantigens for inclusion in a vaccine formulation. Rather than attempt to use CRCL in a vaccine platform that has 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-myeloablative, stem cell transplant (ASCT) proce- dures. After ASCT, a graft vs. tumor (GVT) cascade occurs which has proven ability to kill refractory he- matological malignancies and chemotherapy-resis- tant metastatic solid tumors. However, this beneficial anti-tumor effect is linked to graft vs. host disease (GVHD). The high morbidity and mortality of GVHD severely limits the use of ASCT pro- cedures 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 the engrafted donor cells, the flow instead was engineered to emanate from the host so as to gr94/gp96 hsp70 hsp90 calreticulin AUTHORS 1National Cancer Institute, Bangkok, Thailand; 2Immunovative Therapies, Ltd, Jerusalem, Israel; 3Khon Kaen University Srinagarind Hospital, Khon Kaen, Thailand; 4Immunovative Clinical Research, Inc., Mesa, AZ; 5National Institutes of Health, Nonthaburi, Thailand, 6 [email protected] Wirote Lausoontornsiri1, Michael Har-Noy2,4,6, Jitraporn Wongwiwatcha3, Supaporn Suparak5, Anucha Ahooja3, Miriam Bloch4, Michael Bishop4 and Wattana Sukeepaisarnjaroen3 Abstract # 202873 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. 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 In order to amplify the anti-tumor effects of priming and accelerate the development of anti-tu- mor-specific memory cells in the tumor sites, a biopsy sample of a select tumor lesion is lysed ex-vivo and the endogenous HSP chaperones, including hsp70, hsp90, gr94/gp96 and calreticu- lin are purified from the debris using an isoelectric focusing technique. The purified HSP is called Chaperone-Rich Cell Lysate (CRCL). CRCL is injected Intradermally at the same time as BAG cells. The BAG cells condition the environment with Type 1 cytokines and attract NK cells and memory allo-specific Th1/CTL to the injection site. These cells reject the BAG, which causes the release of endogenous HSP and DAMP. iDC in the skin respond to process the debris from the rejected BAG and engulf the HSP released from BAG and the CRCL. In the presence of DAMP and Type 1 cyto- kines the iDC mature to IL-12+ DC1, traffic to the draining lymph nodes and facilitate develop- ment of activated allo-specific and tumor-specific Th1/CTL, which in turn cause extravasation of memory allo-specific and tumor-specific Th1/CTL to tumor sites. Subsequent injections of CRCL+BAG cause some of the activated allo-specific and tumor-specific Th1/CTL to differentiate into memory cells. Each subsequent injection of CRCL+BAG produces activated allo-specific and tumor-specific Th1/CTL which in turn activate these circulating memory allo-specific and tu- mor-specific Th1/CTL causing them to extravasate to tumor sites. 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. Adaptive Immunity Counter-regulate Immunosuppressive and Immunoavoidance Th2 Mechanisms Adaptive Immunity NK NK CD4 Th1 CD8 CD4 CTL Th1 CTL Th1 CTL CTL Th1 Th1 CTL 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 Allo Antigen Allo Antigen Tumor Antigen Tumor Antigen Type 1 Cytokines Tumor 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. NK i DC i DC DC 1 DC 1 elicit host vs. tumor (HVT) effects linked to a non-toxic host vs. graft (HVG) rejection. 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-ϒ 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-ϒ and express high density CD40L. Upon rejection, BAG release endogenous DAMP. CD40L has the same immune effects as LPS. NOTES: The injection site on days 0 and 3 will be the same and the injection site on days 7 and 10 will be the same, but different from the day 0 and 3 site. The injection site on days 14 and 17 will be the same site as day 0, and the injection site on days 21 and 24 will be the same as day 7, CRCL will be injected into the same ID site ~ 5 minutes after the study drug during the vaccination phase. After protocol completion subjects can continue to receive CRCL alone every 28 days, as tolerated, until vaccine is exhausted or death of the subject, at the discretion of the PI. The therapeutic vaccine design being tested here is unique in that it has both a vaccination component, using patient-derived purified CRCL with BAG, and a method for disrupting the suppression in the tumor microenvironment. The multiple feed-forward waves of activated allo-specific and tumor-specific Th1/CTL that occur after each dosing can act to modulate the tumor microenvironment to a Type 1 environment. Under these Type 1 conditions, immune mediated tumor debulking responses are possible. Exploratory radiological, pathological and biological end-points are being evaluated in longitudinal CT scans, biopsies and serum samples. mRECIST, which evaluates changes in the viable tumor fractions on CT scan, is used as an efficacy end-point. Longitudinal CT scans and biopsies are taken simultaneously. The bi- opsies are evaluated for changes in liver cellular anat- omy, tumor burden, immune cell infiltration, necrosis and fibrosis. PBMC samples are analyzed for biomark- ers, such as HSP70 and IL-6. Phase Dosing Schedule Priming Day 0 ID -1 ml Day 3 ID -1 ml Day 7 ID -1 ml Day 10 ID -1 ml Vaccination Day 14 ID - 1 ml + ID CRCL Day 17 ID - 1 ml + ID CRCL Day 21 ID - 1 ml + ID CRCL Day 24 ID - 1 ml + ID CRCL Activation Day 28 IV - 10 ml Booster Day 56 IV - 5 ml + ID CRCL Day 84 IV - 5 ml + ID CRCL Day 112 IV - 5 ml + ID CRCL Day 140 IV - 5 ml + ID CRCL Day 168 IV - 5 ml + ID CRCL

Upload: others

Post on 24-Sep-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Phase II Trial of a Personalized˜Vaccine for Advanced HCC€¦ · This trial-in-progress is evaluating a new therapeutic cancer vaccine platform in advanced hepatocellular carcinoma

hsp70, hsp90, gr94/gp96 and calreticulin. This Chaperone-Rich

Cell Lysate (CRCL) is quality tested to assure the identity, purity, sterility, low endo-toxin and integrity of the chaperoned tumor anti-gens. The puri�ed HSP are injected intradermally together with BAG cells.

In order to customize an immune response against a patient’s own tumor cells and increase the titers of activated allo-speci�c and tumor-speci�c Th1/CTL in circulation, a biopsy sample of a tumor is collect-ed and sent to Jerusalem for processing. In Jerusalem, the tumors are processed under Good Manufacturing Practices (GMP) to isolate endogenous HSP, including

Phase II Trial of a Personalized Vaccine for Advanced HCC

AbstractBackground: The chronic in�ammation and viral infection associated with HCC combined with liver tolerogenic mechanisms creates a profoundly immunosuppressive microenviron-ment. Modulating the microenvironment with check-point blockade bene�ts ~20% of patients. The non-re-sponders and those with poor liver function represent an unmet medical need. Checkpoint blockade requires ef-fector cells to be resident within tumors. However, the majority of HCC lesions lack in�ltrating e�ector cells. Therefore, a strategy for amplifying the tumor-speci�c immune response, while counteracting the immunosup-pressive mechanisms may provide an improved immuno-therapy. Endogenous heat shock proteins (HSP) chaper-one tumor neoantigens. A process enabling the concen-tration of HSP such as calreticulin, hsp70, hsp90 and gr94/gp96 (CRCL) from tumor biopsy samples has been developed and serves as a source of tumor neoantigen for vaccination. In addition, a bioengineered allogeneic allograft (BAG) derived from healthy blood donors with potent immunomodulatory and counter-regulatory properties has been developed as an adjuvant. The com-bination of CRCL with BAG is being evaluated as a strate-gy to amplify tumor-speci�c immunity and counter-regu-late the immunosuppressive microenvironment in ad-vanced HCC.

Methods: An open-label Phase II clinical trial evaluating the safety and e�cacy of CRCL+BAG vaccine in Child-Pugh A/B ad-vanced or metastatic HCC w/ or w/o Sorafenib is being conducted at Khon Kaen University Medical Center in Thailand . Approximately 6-10 biopsy cores are collected at baseline. The tumor sampe is lysed ex-vivo and the en-dogenous CRCL puri�ed using an isoelectric focusing technique. Subjects are �rst primed with multiple intra-dermal injections of BAG cells alone to increase the titer of allo-speci�c Th1/CTL cells. Next, multiple intradermal injections of BAG+CRCL are administered to elicit in-creased titers of tumor-speci�c Th1/CTL cells. Subsequent intravenous infusions of BAG cells activate allo-speci�c and tumor-speci�c memory cells, enabling their in�ltra-tion into tumor lesions. The BAG allo-rejection response creates a type I ‘cytokine storm’ which serves to down-reg-ulate suppressor circuits. Response is evaluated by mRE-CIST. Longitudinal CT scans with concurrent biopsies as well as multiple collections of PBMC and plasma permits immunomonitoring of these immune mechanisms.

Clinical trial information: NCT02409524

DiscussionCurrent therapies for advanced HCC are marginally e�ective and exacerbate underlying liver disease. The inherent immune tolerance of the liver suppresses cellular immunity and permits HCC progression and spread.

The liver is exposed to abundant xenoantigens from the gut via the portal vein. These xenoantigens tend to polarize the liver microenvironment to Type 2 which induces immune tolerance and recruitment of suppressor cells, such as Treg and MDSC. In addition, the Type 2 conditions upregulate expression of check-point molecules, such programmed death ligand 1 (PD-L1), which contributes to further weaken anti-tu-mor immune responses.

Nonparenchymal cells, including stellate cells, hepatic dendritic cells and liver sinusoidal endothelial cells also can induce tolerance and/or apoptosis of e�ector Th1/CTL cells, further exacerbating the hostile envi-ronment for cellular anti-tumor immune function.

Dosing Schedule

IntroductionThis trial-in-progress is evaluating a new therapeutic cancer vaccine platform in advanced hepatocellular carcinoma (HCC). HCC is one of the most common ma-lignancies worldwide and is endemic in Southeast Asia due to the high incidence of hepatitis B and hep-atitis C. Some of the highest incidence rates occur within the Khon Kaen region of Thailand, which is the site of this study. In Thailand, most HCC patients pres-ent to the clinic with advanced Barcelona Clinic Liver Cancer [BCLC] stage C disease. These patients have limited therapeutic options. Approved therapies including sorafenib, regorafenib and nivolomab have response rates <20% and only a modest bene�t in terms of overall survival. In addition, these treatments are generally too expensive for widescale use in the Thai population.

Since HCC has been shown to be immunogenic and is responsive to checkpoint blockade, therapeutic vacci-nation represents a promising treatment option for these patients. Successful development of a thera-peutic vaccine has potential to increase survival and to have lower cost than current options. However, while past therapeutic vaccine immunotherapy strat-egies against HCC have demonstrated tumor-speci�c T cell responses, these responses were often not robust enough to induce clinical responses.

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-inducing 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-domi-nated immunity both systemically and in the tumor microenvironment; and

(6) immunosuppression due to the immune privi-leged nature of the liver biology.

In chronically in�amed livers, genetic and epigenetic changes underlie oncogenic transformation and pro-duce mutations which can be recognized as neoanti-gens by the immune system. The complete antigenic repertoire of tumor cells is associated with endoge-nous heat shock proteins (HSP). We developed a method of purifying HSP from tumor biopsy samples resulting in a vaccine preparation enriched in hsp70, hsp90, gr94/gp96 and calreticulin (known as chaper-one rich cell lysate or “CRCL”). CRCL provides a source of patient-speci�c tumor neoantigens for inclusion in a vaccine formulation.

Rather than attempt to use CRCL in a vaccine platform that has 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-myeloablative, stem cell transplant (ASCT) proce-dures. After ASCT, a graft vs. tumor (GVT) cascade occurs which has proven ability to kill refractory he-matological malignancies and chemotherapy-resis-tant metastatic solid tumors.

However, this bene�cial anti-tumor e�ect is linked to graft vs. host disease (GVHD). The high morbidity and mortality of GVHD severely limits the use of ASCT pro-cedures 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 the engrafted donor cells, the �ow instead was engineered to emanate from the host so as to

gr94/gp96

hsp70

hsp90

calreticulin

A U T H O R S

1National Cancer Institute, Bangkok, Thailand;

2Immunovative Therapies, Ltd, Jerusalem, Israel;

3Khon Kaen University Srinagarind Hospital, Khon Kaen, Thailand;

4Immunovative Clinical Research, Inc., Mesa, AZ;

5National Institutes of Health, Nonthaburi, Thailand,

6 [email protected]

Wirote Lausoontornsiri1, Michael Har-Noy2,4,6, Jitraporn Wongwiwatcha3, Supaporn Suparak5,

Anucha Ahooja3, Miriam Bloch4, Michael Bishop4 and Wattana Sukeepaisarnjaroen3

Abstract # 202873

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. Vaccination Phase

The 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�cationIn order to amplify the anti-tumor e�ects of priming and accelerate the development of anti-tu-mor-speci�c memory cells in the tumor sites, a biopsy sample of a select tumor lesion is lysed ex-vivo and the endogenous HSP chaperones, including hsp70, hsp90, gr94/gp96 and calreticu-lin are puri�ed from the debris using an isoelectric focusing technique. The puri�ed HSP is called Chaperone-Rich Cell Lysate (CRCL). CRCL is injected Intradermally at the same time as BAG cells. The BAG cells condition the environment with Type 1 cytokines and attract NK cells and memory allo-speci�c Th1/CTL to the injection site. These cells reject the BAG, which causes the release of endogenous HSP and DAMP. iDC in the skin respond to process the debris from the rejected BAG and engulf the HSP released from BAG and the CRCL. In the presence of DAMP and Type 1 cyto-kines the iDC mature to IL-12+ DC1, tra�c to the draining lymph nodes and facilitate develop-ment of activated allo-speci�c and tumor-speci�c Th1/CTL, which in turn cause extravasation of memory allo-speci�c and tumor-speci�c Th1/CTL to tumor sites. Subsequent injections of CRCL+BAG cause some of the activated allo-speci�c and tumor-speci�c Th1/CTL to di�erentiate into memory cells. Each subsequent injection of CRCL+BAG produces activated allo-speci�c and tumor-speci�c Th1/CTL which in turn activate these circulating memory allo-speci�c and tu-mor-speci�c Th1/CTL causing them to extravasate to tumor sites.

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.

Adaptive Immunity

Counter-regulate Immunosuppressive and Immunoavoidance Th2 Mechanisms

Adaptive Immunity

NK

NK

CD4

Th1

CD8

CD4 CTLTh1

CTLTh1

CTL

CTLTh1Th1 CTL

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

Allo Antigen

Allo AntigenTumor

Antigen

Tumor Antigen

Type 1 Cytokines

Tumor

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.

NK

iDC

iDC DC1

DC1

elicit host vs. tumor (HVT) e�ects linked to a non-toxic host vs. graft (HVG) rejection.

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-ϒ 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-ϒ and express high density CD40L. Upon rejection, BAG release endogenous DAMP. CD40L has the same immune e�ects as LPS.

NOTES:

The injection site on days 0 and 3 will be the same and the injection site on days 7 and 10 will be the same, but di�erent from the day 0 and 3 site.

The injection site on days 14 and 17 will be the same site as day 0, and the injection site on days 21 and 24 will be the same as day 7, CRCL will be injected into the same ID site ~ 5 minutes after the study drug during the vaccination phase.

After protocol completion subjects can continue to receive CRCL alone every 28 days, as tolerated, until vaccine is exhausted or death of the subject, at the discretion of the PI.

The therapeutic vaccine design being tested here is unique in that it has both a vaccination component, using patient-derived puri�ed CRCL with BAG, and a method for disrupting the suppression in the tumor microenvironment. The multiple feed-forward waves of activated allo-speci�c and tumor-speci�c Th1/CTL that occur after each dosing can act to modulate the tumor microenvironment to a Type 1 environment. Under these Type 1 conditions, immune mediated tumor debulking responses are possible.

Exploratory radiological, pathological and biological end-points are being evaluated in longitudinal CT scans, biopsies and serum samples. mRECIST, which evaluates changes in the viable tumor fractions on CT scan, is used as an e�cacy end-point. Longitudinal CT scans and biopsies are taken simultaneously. The bi-opsies are evaluated for changes in liver cellular anat-omy, tumor burden, immune cell in�ltration, necrosis and �brosis. PBMC samples are analyzed for biomark-ers, such as HSP70 and IL-6.

Phase Dosing Schedule

Priming

Day 0 ID -1 ml

Day 3 ID -1 ml

Day 7 ID -1 ml

Day 10 ID -1 ml

Vaccination

Day 14 ID - 1 ml + ID CRCL

Day 17 ID - 1 ml + ID CRCL

Day 21 ID - 1 ml + ID CRCL

Day 24 ID - 1 ml + ID CRCL

Activation Day 28 IV - 10 ml

Booster

Day 56 IV - 5 ml + ID CRCL

Day 84 IV - 5 ml + ID CRCL

Day 112 IV - 5 ml + ID CRCL

Day 140 IV - 5 ml + ID CRCL

Day 168 IV - 5 ml + ID CRCL