A CD40 Agonist and PD-1 Antagonist Antibody Reprogram the Microenvironment of Nonimmunogenic Tumors to Allow T-cell–Mediated Anticancer Activity (original) (raw)
Related papers
Agonistic CD40 mAb-Driven IL12 Reverses Resistance to Anti-PD1 in a T-cell–Rich Tumor
Cancer Research, 2016
The durability and efficacy of anti-human PD1 monoclonal antibodies (PD1 mAb) vary across different malignancies. Although an absence of tumor-infiltrating cytotoxic T lymphocytes has been identified as a cause for resistance to PD1 mAb, the presence of intratumor exhausted PD1hi T cells also contributes to insensitivity to this immune checkpoint therapy. In this study, we used mouse tumor models of PD1 mAb resistance that harbored PD1hi T cells and flow cytometry analysis of tumor-infiltrating leukocytes immediately post-therapy as a screening platform to identify agents that could resensitize T cells to PD1 blockade. We showed that an agonistic anti-CD40 mAb converted PD1hi T cells into PD1lo T cells, reversing phenotypic T-cell exhaustion and allowing the anti-PD1 refractory tumors to respond to anti-PD1 therapy. PD1 downmodulation by anti-CD40 mAb relied upon IL12 but not IL23, CD80/CD86/CD28, or CD70/CD27. Consistent with a role for regulatory T cells (Treg) in promoting T-cell...
Proceedings of the National Academy of Sciences, 2002
Adequate spontaneous activation of tumor-specific T lymphocytes in tumor-bearing hosts is rare, despite the expression of tumor antigens that are potentially highly immunogenic. For example, failure of the immune system to raise competent responses against established tumors expressing the human adenovirus E1A-antigen allows this tumor to grow in immunocompetent mice. We show that systemic in vivo administration of agonistic anti-CD40 antibodies into tumor-bearing mice results in tumor eradication mediated by CD8 ؉ T cells. Treatment resulted in a strong expansion and systemic accumulation of E1A-specific CTL and depended on CD40 expression on host cells, as the tumor was CD40 ؊ , and therapy failed in CD40-deficient mice. Local intratumoral administration of anti-CD40 mAb is equally effective in licensing strong, systemic CTL immunity, resulting in the clearance of distant tumor nodules. Our data indicate that the immune response after cancerhost interactions can be directed toward competence, leading to the cure of established tumors merely by delivery of a CD40dependent ''license to kill'' signal.
Journal for ImmunoTherapy of Cancer
BackgroundPancreatic ductal adenocarcinoma (PDAC) is notoriously resistant to treatment including checkpoint-blockade immunotherapy. We hypothesized that a bimodal treatment approach consisting of dendritic cell (DC) vaccination to prime tumor-specific T cells, and a strategy to reprogram the desmoplastic tumor microenvironment (TME) would be needed to break tolerance to these pancreatic cancers. As a proof-of-concept, we investigated the efficacy of combined DC vaccination with CD40-agonistic antibodies in a poorly immunogenic murine model of PDAC. Based on the rationale that mesothelioma and pancreatic cancer share a number of tumor associated antigens, the DCs were loaded with either pancreatic or mesothelioma tumor lysates.MethodsImmune-competent mice with subcutaneously or orthotopically growing KrasG12D/+;Trp53R172H/+;Pdx-1-Cre (KPC) PDAC tumors were vaccinated with syngeneic bone marrow-derived DCs loaded with either pancreatic cancer (KPC) or mesothelioma (AE17) lysate and c...
Optimized antiangiogenic reprogramming of the tumor microenvironment potentiates CD40 immunotherapy
Proceedings of the National Academy of Sciences
Cancer immunotherapies are increasingly combined with targeted therapies to improve therapeutic outcomes. We show that combination of agonistic anti-CD40 with antiangiogenic antibodies targeting 2 proangiogenic factors, vascular endothelial growth factor A (VEGFA) and angiopoietin 2 (Ang2/ANGPT2), induces pleiotropic immune mechanisms that facilitate tumor rejection in several tumor models. On the one hand, VEGFA/Ang2 blockade induced regression of the tumor microvasculature while decreasing the proportion of nonperfused vessels and reducing leakiness of the remaining vessels. On the other hand, both anti-VEGFA/Ang2 and anti-CD40 independently promoted proinflammatory macrophage skewing and increased dendritic cell activation in the tumor microenvironment, which were further amplified upon combination of the 2 treatments. Finally, combined therapy provoked brisk infiltration and intratumoral redistribution of cytotoxic CD8+ T cells in the tumors, which was mainly driven by Ang2 bloc...
In situ vaccination with defined factors overcomes T cell exhaustion in distant tumors
Journal of Clinical Investigation
Irreversible T cell exhaustion limits the efficacy of programmed cell death 1 (PD-1) blockade. We observed that dual CD40-TLR4 stimulation within a single tumor restored PD-1 sensitivity and that this regimen triggered a systemic tumor-specific CD8 + T cell response. This approach effectively treated established tumors in diverse syngeneic cancer models, and the systemic effect was dependent on the injected tumor, indicating that treated tumors were converted into necessary components of this therapy. Strikingly, this approach was associated with the absence of exhausted PD-1 hi T cells in treated and distant tumors, while sparing the intervening draining lymph node and spleen. Furthermore, patients with transcription changes like those induced by this therapy experienced improved progression-free survival with anti-PD-1 treatment. Dual CD40-TLR4 activation within a single tumor is thus an approach for overcoming resistance to PD-1 blockade that is unique in its ability to cause the loss of exhausted T cells within tumors while sparing nonmalignant tissues.
Theranostics
Background: Advanced stage cancers with a suppressive tumor microenvironment (TME) are often refractory to immune checkpoint inhibitor (ICI) therapy. Recent studies have shown that focused ultrasound (FUS) TME-modulation can synergize ICI therapy, but enhancing survival outcomes in poorly immunogenic tumors remains challenging. Here, we investigated the role of focused ultrasound based boiling histotripsy (HT) and in-situ anti-CD40 agonist antibody (αCD40) combinatorial therapy in enhancing therapeutic efficacy against ICI refractory murine melanoma. Methods: Unilateral and bilateral large (~330-400 mm 3) poorly immunogenic B16F10 melanoma tumors were established in the flank regions of mice. Tumors were exposed to single local HT followed by an in-situ administration of αCD40 (HT+ αCD40: HT40). Inflammatory signatures post treatment were assessed using pan-cancer immune profiling and flow cytometry. The ability of HT40 ± ICI to enhance local and systemic effects was determined by immunological characterization of the harvested tissues, and by tumor growth delay of local and distant untreated tumors 4-6 weeks post treatment. Results: Immune profiling revealed that HT40 upregulated a variety of inflammatory markers in the tumors. Immunologically, HT40 treated tumors showed an increased population of granzyme B+ expressing functional CD8+ T cells (~4-fold) as well as an increased M1 to M2 macrophage ratio (~2-3-fold) and CD8+ T: regulatory T cell ratio (~5-fold) compared to the untreated control. Systemically, the proliferation rates of the melanoma-specific memory T cell population were significantly enhanced by HT40 treatment. Finally, the combination of HT40 and ICI therapy (anti-CTLA-4 and anti-PD-L1) caused superior inhibition of distant untreated tumors, and prolonged survival rates compared to the control. Conclusions: Data suggest that HT40 reprograms immunologically cold tumors and sensitizes them to ICI therapy. This approach may be clinically useful for treating advanced stage melanoma cancers.
CD40 and CD70 co-stimulate a potent in vivo anti-tumor T cell response
Immunology Letters, 1997
In several studies, CD80, a potent co-stimulatory molecule, has been reported to be responsible for the induction of CD8' antitumor T cell responses by CD80-transfected tumor cells. However, expression of CD80 by tumors not always ensures generation of a T cell-mediated antitumor response. Variables such as the inherent immunogenicity of a tumor and its major histocompatibility complex (MHC) expression status affect the efficacy of this approach. Therefore, in this study two other co-stimulatory ligands, CD40 and CD70, have been investigated for their ability to co-stimulate antitumor responses. The efficacy of CD40 and CD70 is compared with that of CD80, with respect to CD4 and CD8 T cell co-stimulatory capacity in vitro and their ability to induce in vivo antitumor responses. Furthermore, CD40 and CD70 are tested for their capacity to induce a long-lived memory response in vivo, as defined both by induction of tumor-specific cytotoxic T lymphocytes (CTLs) and rejection of wild-type tumor cells. It was found that, despite the fact that CD40 predominantly stimulates CD4 T cells, CD4O-transfected MHC class 11-negative P8 15 tumor cells become highly immunogenic and induce long-lasting memory tumor-specific CTLs in vivo. Furthermore, CD40 and CD70 emerge as powerful and even superior alternatives to CD80 for improving tumor immunogenicity in vivo. While the mechanisms by which they do so remain to be defined, these findings suggest additional strategies for immunotherapy.
BMC cancer, 2017
CD40 signalling can synergise with chemotherapy in preclinical cancer models, and early clinical studies are promising. We set out to define the immunological changes associated with this therapeutic combination to identify biomarkers for a response to the therapy. Here, we present serial immunomonitoring examining dendritic cell and T cell subpopulations over sequential courses of chemoimmunotherapy. Fifteen patients with mesothelioma received up to six 21-day cycles of pemetrexed plus cisplatin chemotherapy and anti-CD40 (CP-870,893). Peripheral blood was collected weekly, and analysed by flow cytometry. Longitudinal immunophenotyping data was analysed by linear mixed modelling, allowing for variation between patients. Exploratory analyses testing for any correlation between overall survival and immunophenotyping data were undertaken up to the third cycle of treatment. Large statistically significant cyclical variations in the proportions of BDCA-1+, BDCA-2+ and BDCA-3+ dendritic ...
Research Square (Research Square), 2024
While CD40 agonism is an attractive approach for activating antigen-presenting cells and initiating antitumor responses, previous attempts have encountered limited clinical efficacy coupled with toxicity. We previously demonstrated that interactions between the antibody Fc domain and the inhibitory receptor FcgRIIB are critical for enhanced antitumor activity. Here, we present the results of a phase 1 study on intratumoral administration of an anti-CD40 agonistic antibody (2141-V11) Fc-engineered to enhance FcgRIIB binding. Primary endpoints included safety, maximum tolerated dose (MTD), and recommended phase 2 dose. Secondary objectives included preliminary clinical activity and correlative studies from biospecimens. 2141-V11 was well-tolerated without dose-limiting toxicities and MTD was not reached. In ten evaluable patients with metastatic cancer, the overall response rate was 20%, with complete responses in two patients (melanoma and breast carcinoma) and stable disease in six patients. 2141-V11 induced tumor regression in injected and non-injected lesions, with increased leukocyte infiltration and tertiary lymphoid structures (TLS) formation in post-treatment biopsies. In a humanized mouse model for CD40 and FcgRs, 2141-V11 induced TLS formation in mice bearing orthotopic breast carcinoma, correlating with local and abscopal antitumor effects, systemic immune activation, and immune memory. These findings support the safety and efficacy of 2141-V11, warranting phase 2 studies and suggesting a unique mechanism of action for this Fc-enhanced immunotherapy (NCT04059588). Main 41 Blocking inhibitory immune receptors with anti-PD-1/PD-L1 and anti-CTLA-4 antibodies (Abs) is now established as an effective therapeutic strategy for several malignancies 1-3. However, the benefit of these therapies remains limited to a small subset of patients. Activating the immune system through stimulatory pathways represents a promising alternative and complementary approach. In particular, the stimulatory receptor CD40 plays a central role in promoting antitumor immunity and developing tumor-specific T-cell responses 4. CD40 is a member of the tumor necrosis receptor superfamily, expressed predominantly on antigen-presenting cells (APCs) such as dendritic cells (DCs), B-cells and macrophages. When CD40 binds to its ligand, CD40 ligand (CD154), it trimerizes to initiate downstream signaling to activate APCs 5 (Scheme 1a). This activation is critical for promoting antigen presentation by DCs, which is crucial step to induce effective T-cell responses 6. Engagement of CD40 can be achieved not only through its cognate ligand but also through agonistic Abs, which promote APC maturation and activation, resulting in T-cell responses and elimination of tumor cells 7. Several first-generation CD40 agonist Abs were developed and tested in patients with cancer. However, they demonstrated dose-limiting systemic toxicities (mainly thrombocytopenia and transaminitis) and had limited clinical benefit 8-14. We have previously demonstrated that the in vivo activity of agonistic anti-CD40 Abs relies on interactions between the Ab fragment crystallizable (Fc) domain and the inhibitory Fc-gamma receptor FcgIIB, resulting in optimal trimerization and agonistic signaling 15-18 (Scheme 1b). Based on these findings, we engineered 2141-V11, a human IgG anti-CD40 agonistic Ab, which had previously been evaluated in several phase 1 studies as an IgG2 antibody (clone CP-870,893) 19,20 , with point mutations in its Fc portion to selectively increase binding affinity to FcgRIIB 15. Using an immunocompetent mouse model humanized for both CD40 and all human FcgRs (hCD40/hFcgR mice), we demonstrated that 2141-64 V11 has enhanced immune stimulatory activity in vivo, resulting in enhanced DC activation and 65 CD8 anti-tumor immunity when compared to other clinical CD40 Abs lacking selective FcgRIIB binding across several tumor models 15. Studies in our hCD40/hFcgR mouse model also demonstrated that systemic administration of 2141-V11 was associated with increased levels of toxicity (thrombocytopenia and transaminitis) 18. Importantly, these toxicities were circumvented by intratumoral (IT) administration, allowing for effective in situ vaccination effect with systemic antitumor activity and minimal toxicity. These studies provided the mechanistic foundation for our first-inhuman investigatorinitiated study (NCT04059588), assessing the safety and tolerability of IT delivery of 2141-V11 in patients with solid tumors locally advanced or metastatic to the skin amenable to IT injection. By optimizing the Fc portion of the CD40 agonistic Ab, and by switching the administration route from systemic to IT, we have addressed the challenges that have led to poor performance of prior anti-CD40 agonistic Abs. The primary endpoint was to determine the maximum tolerated dose (MTD) and the recommended phase 2 dose (RP2D) of 2141-V11. Secondary objectives include evaluating pharmacokinetics and preliminary clinical activity. Immune profiling of biospecimens obtained from patients that achieved complete responses (CR) and in vivo mechanistic experiments using our hCD40/hFcgR mice complement these studies, providing further characterization of the mechanism of action of 2141-V11. Results Preclinical toxicology and determination of human dosing of 2141-V11 Several pre-clinical assessments were performed to identify a safe initial dose for human trials, aiming to minimize toxicity risk and limit the exposure of subjects to ineffective doses. Pharmacokinetics and toxicology studies were performed in non-human primates (NHP) 88 receiving subcutaneous (SC) or intravenous (IV) 2141-V11 (Extended data Fig. 1). 2141-V11 89 was detected in serum samples from NHP, with concentration versus time profiles consistent with 90 the route of administration, and peak serum concentrations between and 72 hours (Extended 91