scholarly journals Overcoming primary and acquired resistance to anti-PD-L1 therapy by induction and activation of tumor-residing cDC1s

2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Takaaki Oba ◽  
Mark D. Long ◽  
Tibor Keler ◽  
Henry C. Marsh ◽  
Hans Minderman ◽  
...  

Abstract The ability of cancer cells to ensure T-cell exclusion from the tumor microenvironment is a significant mechanism of resistance to anti-PD-1/PD-L1 therapy. Evidence indicates crucial roles of Batf3-dependent conventional type-1 dendritic cells (cDC1s) for inducing antitumor T-cell immunity; however, strategies to maximize cDC1 engagement remain elusive. Here, using multiple orthotopic tumor mouse models resistant to anti-PD-L1-therapy, we are testing the hypothesis that in situ induction and activation of tumor-residing cDC1s overcomes poor T-cell infiltration. In situ immunomodulation with Flt3L, radiotherapy, and TLR3/CD40 stimulation induces an influx of stem-like Tcf1+ Slamf6+ CD8+ T cells, triggers regression not only of primary, but also untreated distant tumors, and renders tumors responsive to anti-PD-L1 therapy. Furthermore, serial in situ immunomodulation (ISIM) reshapes repertoires of intratumoral T cells, overcomes acquired resistance to anti-PD-L1 therapy, and establishes tumor-specific immunological memory. These findings provide new insights into cDC1 biology as a critical determinant to overcome mechanisms of intratumoral T-cell exclusion.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1581-1581
Author(s):  
Roch Houot ◽  
R. Levy

Abstract Introduction: We have previously shown that intratumoral injection of CpG oligodeoxynucleotide plus systemic chemotherapy can induce T cell immunity against lymphoma and serve as a therapeutic vaccine to cure tumors in a murine lymphoma model (Li et al., J Immunol. 2007). CpG is a Toll-like receptor 9 agonist capable of activating tumor-infiltrating dendritic cells and/or tumor B cells to force tumor antigen presentation in situ when injected intratumorally. Here, we demonstrate that antibody-mediated modulation of T cells enhances the efficacy of CpG vaccination, thereby eliminating the need for chemotherapy. Materials and Methods: Mice were inoculated s.c. with A20 lymphoma tumor cells at two sites (right and left abdomen). Only one site was injected with CpG allowing us to evaluate the systemic anti-tumor response at the distant site. Treatment started when tumors became palpable. CpG was administered intratumorally. T cell modulation was accomplished using systemic (i.p.) administration of monoclonal antibodies against T cell targets: - Regulatory T cells were depleted using anti-Folate Receptor 4 (FR4) antibody or functionally blocked using anti-GITR antibody, - Effector T cells were stimulated using anti-OX40 antibody (to trigger their costimulatory molecule) or anti-CTLA4 antibody (to block inhibitory signals). Results: Treatment with intratumoral injection of CpG alone did not cure any mice. Treatment with CpG and a single antibody (anti-OX40, anti-CTLA4, anti-FR4, or anti-GITR) cured 20–30% of mice. Interestingly, some combinations of antibodies (anti-OX40+anti-CTLA4, anti-OX40+anti-FR4, anti-CTLA4+anti-GITR) potentiated T cell modulation and further enhanced the efficacy of CpG vaccination. In particular, the combination of anti-OX40 and anti-CTLA4 appeared to be especially potent when combined with intratumoral CpG. Indeed, this combination (CpG+anti-OX40+anti-CTLA4) induced anti-tumor T cells capable of secreting IFN-γ in response to overnight culture with A20 tumor cells; it cured more than 80% of mice bearing large and systemic lymphoma tumors without the need for chemotherapy (effective therapy required both CD4 and CD8 T cells); finally, this therapy produced high numbers of anti-tumor memory T cells and provided long-lasting immunity against tumor re-challenge. Conclusions: Our results show that antibody-mediated T cell modulation greatly enhances the therapeutic efficacy of intratumoral vaccination with CpG. Importantly, we show that T cell modulation can be potentiated by appropriate combinations of antibodies against T cell targets. As these reagents (CpG, anti-OX40 and anti-CTLA4 notably) are becoming available for human clinical trials, the combination of intratumoral CpG and immunomodulatory T cell antibodies holds promise for therapeutic vaccination against lymphoma.


Blood ◽  
2009 ◽  
Vol 113 (1) ◽  
pp. 85-94 ◽  
Author(s):  
Joshua D. Brody ◽  
Matthew J. Goldstein ◽  
Debra K. Czerwinski ◽  
Ronald Levy

Abstract Ex vivo–expanded tumor-infiltrating lymphocytes infused into lymphodepleted recipients has clear antitumor efficacy. More practical sources of such antitumor lymphocytes would broaden the application of this approach. Previously, we described an in situ vaccination combining chemotherapy with intratumoral injection of CpG-enriched oligonucleotides, which induced T-cell immunity against established lymphoma. An ongoing clinical trial of this maneuver has demonstrated clinical responses in lymphoma patients. Here, we use this vaccine maneuver to generate immune cells for transfer into irradiated, syngeneic recipients. Transferred tumor-specific T-effector (Teff) cells preferentially expanded, increasing the Teff/T-regulatory (Treg) ratio in these “immunotransplantation” recipients and curing large and metastatic tumors. Donor T cells were necessary for tumor protection, and CD8 T-cell immune responses were enhanced by posttransplantation booster vaccination. Hematopoietic stem cell transplantation is a standard therapy for lymphoma. Therefore, in situ tumor vaccination followed by immunotransplantation of harvested tumor-specific T cells could be directly tested in clinical trials to treat otherwise resistant malignancies.


2022 ◽  
Vol 219 (2) ◽  
Author(s):  
Dimitra Kerdidani ◽  
Emmanouil Aerakis ◽  
Kleio-Maria Verrou ◽  
Ilias Angelidis ◽  
Katerina Douka ◽  
...  

A key unknown of the functional space in tumor immunity is whether CD4 T cells depend on intratumoral MHCII cancer antigen recognition. MHCII-expressing, antigen-presenting cancer-associated fibroblasts (apCAFs) have been found in breast and pancreatic tumors and are considered to be immunosuppressive. This analysis shows that antigen-presenting fibroblasts are frequent in human lung non-small cell carcinomas, where they seem to actively promote rather than suppress MHCII immunity. Lung apCAFs directly activated the TCRs of effector CD4 T cells and at the same time produced C1q, which acted on T cell C1qbp to rescue them from apoptosis. Fibroblast-specific MHCII or C1q deletion impaired CD4 T cell immunity and accelerated tumor growth, while inducing C1qbp in adoptively transferred CD4 T cells expanded their numbers and reduced tumors. Collectively, we have characterized in the lungs a subset of antigen-presenting fibroblasts with tumor-suppressive properties and propose that cancer immunotherapies might be strongly dependent on in situ MHCII antigen presentation.


2021 ◽  
Vol 11 ◽  
Author(s):  
Matin Dokht Ashoori ◽  
Kensuke Suzuki ◽  
Yosuke Tokumaru ◽  
Naoko Ikuta ◽  
Masaki Tajima ◽  
...  

Blockade of PD-1, an indispensable physiological immunoregulatory mechanism, enhances immune activities and is widely used in the immunotherapy of cancer. This treatment often accompanies inflammatory complication called immune-related adverse events (irAE), most frequently in the skin. To analyze how skin inflammation develops by the blockade of PD-1-dependent immunoregulation, we studied the exacerbation of oxazolone-induced contact hypersensitivity by PD-L1 blockade. The inactivation of PD-1 signaling enhanced swelling of the skin with massive CD8+ T cell infiltration. Among PD-1-expressing cells, T cells were the predominant targets of anti-PD-L1 mAb treatment since PD-L1 blockade did not affect skin inflammation in RAG2-/- mice. PD-L1 blockade during immunization with oxazolone significantly promoted the development of hapten-reactive T cells in the draining lymph nodes. The enhancement of local CD8+ T cell-dominant immune responses by PD-L1 blockade was correlated with the upregulation of CXCL9 and CXCL10. Challenges with a low dose of oxazolone did not demonstrate any significant dermatitis; however, the influence of PD-L1 blockade on T cell immunity was strong enough to cause the emergence of notable dermatitis in this suboptimal dosing, suggesting its relevance to dermal irAE development. In the low-dose setting, the blockade of CXCR3, receptor of CXCL9/10, prevented the induction of T cell-dominant inflammation by anti-PD-L1 mAb. This experimental approach reproduced CD8+ T cell-dominant form of cutaneous inflammation by the blockade of PD-L1 that has been observed in dermal irAE in human patients.


2017 ◽  
Vol 26 (1) ◽  
pp. 1-9
Author(s):  
Cornelis R. Van Der Torren ◽  
Jessica S. Suwandi ◽  
Dahae Lee ◽  
Ernst-Jan T. Van't Wout ◽  
Gaby Duinkerken ◽  
...  

Transplantation of islet allografts into type 1 diabetic recipients usually requires multiple pancreas donors to achieve insulin independence. This adds to the challenges of immunological monitoring of islet transplantation currently relying on surrogate immune markers in peripheral blood. We investigated donor origin and infiltration of islets transplanted in the liver of a T1D patient who died of hemorrhagic stroke 4 months after successful transplantation with two intraportal islet grafts combining six donors. Immunohistological staining for donor HLA using a unique panel of human monoclonal HLA-specific alloantibodies was performed on liver cryosections after validation on cryopreserved kidney, liver, and pancreas and compared with auto- and alloreactive T-cell immunity in peripheral blood. HLA-specific staining intensity and signal-to-noise ratio varied between tissues from very strong on kidney glomeruli, less in liver, kidney tubuli, and endocrine pancreas to least in exocrine pancreas, complicating the staining of inflamed islets in an HLA-disparate liver. Nonetheless, five islets from different liver lobes could be attributed to donors 1, 2, and 5 by staining patterns with multiple HLA types. All islets showed infiltration with CD8+ cytotoxic T cells that was mirrored by progressive alloreactive responses in peripheral blood mononuclear cells (PBMCs) to donors 1, 2, and 5 after transplantation. Stably low rates of peripheral islet autoreactive T-cell responses after islet infusion fit with a complete HLA mismatch between grafts and recipient and exclude the possibility that the islet-infiltrating CD8 T cells were autoreactive. HLA-specific immunohistochemistry can identify donor origin in situ and differentiate graft dysfunction and immunological destruction.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. SCI-12-SCI-12
Author(s):  
Nicholas Restifo

Abstract T lymphocytes are at the very center of the rapidly expanding universe of cancer immunotherapy. We now know that each T cell clonotype can in fact contain a stem cell-like system, which is capable of self-renewal and differentiation. We have described the identification of human T memory stem cells (TSCM). TSCM maintain robust replicative capacity and are multipotent in their ability to give rise to cytolytic effector and memory progeny. Stem-cell-like T cells are epigenetically distinct. From their poised epigenetic and metabolic states, TSCM efficiently can give rise to cells with a multiplicity of identities required for successful immune responses, and collectively provide protection against infections and cancer. We have visualized the chromatin states of TSCM on a whole genome level. We have found that the transcription factor Bach2 is a repressor of T cell receptor-driven programs. These findings go a long way in identifying a molecular mechanism that enables T cells to maintain their stem cell-like state. We have direct evidence that sufficient T cell activation can overwhelm the repressive influence of Bach2 through the action of the Akt kinase. Akt mediates the core events of T cell differentiation and it is possible to selectively block the Akt pathway with an allosteric inhibitor enabling the creation of long-lived, less-differentiated T cells that are much more effective therapeutically in a mouse model of established tumor. These findings form the basis for a novel immunometabolomic approach to improve cell-intrinsic features of therapeutic T cells in the clinic. Finally, we will describe how even the most effective T cells can be thwarted by mechanisms in place in the tumor microenvironment. Our recent and unpublished findings involve two elements from the periodic table - potassium and oxygen - that can be immunosuppressive under certain physiologic conditions. We have also described an internal T cell checkpoint mechanism called Cish. We furthermore seek to find ways of overcoming the powerful physiology that can enable tumors to grow and kill their hosts. These efforts can produce T cells that are capable of increased anti-tumor efficacy. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Tianyang Mao ◽  
Eric Song ◽  
Akiko Iwasaki

AbstractCD8+ T cells are required for effective anti-PD-1 (αPD-1) cancer immunotherapy. Type 1 conventional dendritic cells (cDC1s) bearing XCR1 critically mediate the initiation of protective anti-tumor CD8+ T cell responses in mice and humans. However, whether cDC1s contribute to evoking the effector function of CD8+ T cells during αPD-1 antibody therapy remains unclear. Here, by deleting cDC1s at the effector phase of αPD-1 therapy, we identify these cells as a crucial innate determinant for effective αPD-1 immunotherapy. αPD-1 treatment unleashed cDC1s to promote anti-tumor CD8+ T cell immunity, through the expansion of TCF1+ precursors and generation of TIM3+ terminally differentiated effectors. Furthermore, tumor cDC1 abundance was predictive of enhanced CD8+ T cell infiltration, higher survival, and improved clinical responses to αPD-1 therapy in human cancer patients. Together, this study reveals the requirement for cDC1s in PD-1 blockade therapy, through their ability to elicit CD8+ T cell effector responses that mediate tumor control, and highlight cDC1s as an attractive cellular target to be harnessed for novel immunotherapeutics.


2003 ◽  
Vol 198 (2) ◽  
pp. 267-279 ◽  
Author(s):  
Shin-ichiro Fujii ◽  
Kanako Shimizu ◽  
Caroline Smith ◽  
Laura Bonifaz ◽  
Ralph M. Steinman

The maturation of dendritic cells (DCs) allows these antigen-presenting cells to initiate immunity. We pursued this concept in situ by studying the adjuvant action of α-galactosylceramide (αGalCer) in mice. A single i.v. injection of glycolipid induced the full maturation of splenic DCs, beginning within 4 h. Maturation was manifest by marked increases in costimulator and major histocompatibility complex class II expression, interferon (IFN)-γ production, and stimulation of the mixed leukocyte reaction. These changes were not induced directly by αGalCer but required natural killer T (NKT) cells acting independently of the MyD88 adaptor protein. To establish that DC maturation was responsible for the adjuvant role of αGalCer, mice were given αGalCer together with soluble or cell-associated ovalbumin antigen. Th1 type CD4+ and CD8+ T cell responses developed, and the mice became resistant to challenge with ovalbumin-expressing tumor. DCs from mice given ovalbumin plus adjuvant, but not the non-DCs, stimulated ovalbumin-specific proliferative responses and importantly, induced antigen-specific, IFN-γ producing, CD4+ and CD8+ T cells upon transfer into naive animals. In the latter instance, immune priming did not require further exposure to ovalbumin, αGalCer, NKT, or NK cells. Therefore a single dose of αGalCer i.v. rapidly stimulates the full maturation of DCs in situ, and this accounts for the induction of combined Th1 CD4+ and CD8+ T cell immunity to a coadministered protein.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A493-A493
Author(s):  
Takaaki Oba ◽  
Mark Long ◽  
Tibor Keler ◽  
Henry Marsh ◽  
Hans Minderman ◽  
...  

BackgroundThe ability of cancer cells to ensure T-cell exclusion from the tumor microenvironment (TME) is a significant mechanism of resistance to anti-PD-1/PD-L1 therapy. Evidence indicates crucial roles of Batf3-dependent conventional type 1 dendritic cells (cDC1s) for inducing antitumor T-cell immunity. However, strategies to maximize the engagement of cDC1s into such ‘immune cold tumors‘ remain elusive. Using multiple syngeneic orthotopic mouse models of tumors resistant to anti-PD-L1-therapy, we hypothesized that in situ induction and activation of tumor-residing cDC1s overcomes poor T-cell infiltration.MethodsWe utilized three mouse non-T cell-inflamed tumor models that are refractory to anti-PD-L1 therapy (AT-3, B16 and 4T1), and evaluated the efficacy of the combinatorial therapeutic regimen, in situ immunomodulation (ISIM) comprised of intratumoral administration of Fms-like tyrosine kinase 3 receptor ligand (Flt3L) to mobilize cDC1s to the TME, local radiotherapy (RT) to promote immunogenic death of cancer cells and maturation of DCs, and peritumoral CD40/toll-like receptor 3 (TLR3) agonists administration to activate antigen-loaded cDC1s for priming and expansion of tumor-specific CD8+ T cells.ResultsIntratumoral administration of Flt3L increased the number of CD103+ DCs in the TME, and RT induced upregulation of CD40 and CD86 in the tumor-residing CD103+ DCs. In situ CD40/TLR3 stimulation facilitated trafficking of CD103+ DCs carrying tumor-associated antigens (TAA) to the tumor draining LN (TdLN), and generation of tumor-specific CD8+ T cells in TdLNs, indicating cross-presentation of TAA. Consequently, ISIM triggered infiltration of tumor-specific stem-like Tcf1+CD8+ T cells into the TME, mediated rapid regression of untreated distant and primary tumors, and rendered poorly T cell-infiltrated tumors responsive to PD-L1 blockade in multiple mouse tumor models. Moreover, T-cell receptor (TCR) sequencing of TILs revealed that ISIM facilitated the infiltration of novel clones in the TME. Importantly, serial ISIM further reshaped the TCR repertoires in the TME which had been destined to become resistant to anti-PD-L1 therapy, and rendered tumors continuously responsive to anti-PD-L1 therapy, resulting in durable complete responses and establishment of tumor-specific immunological memory.ConclusionsTaken together, ISIM not only increased CD8+ T-cell infiltration but also reshaped the intratumoral TCR repertoires. These findings provide insights into the utility of an in situ combinatorial immunotherapeutic regimen for overcoming resistance to anti-PD-L1 therapy due to tumor-mediated mechanisms of immune cell exclusion.AcknowledgementsWe thank the NIH Tetramer Core Facility (contract HHSN272201300006C) for provision of MHC-I tetramers, This work was supported by National Cancer Institute (NCI) grant P30CA016056 involving the use of Roswell Park’s Flow and Image Cytometry, Pathology Network, Bioinformatics, and Mouse Tumor Model Shared Resource. This work was supported by institutional funds from Roswell Park Comprehensive Cancer Center, the Melanoma Research Alliance (F. Ito), Uehara Memorial Foundation (T. Oba), National Cancer Institute (NCI) grant, K08CA197966 (F. Ito), R50CA211108 (H. Minderman), U24CA232979 (S. Liu) and R01CA172105 (S. Abrams).


Sign in / Sign up

Export Citation Format

Share Document