Journal Articles

IL-12 drives the expression of the inhibitory receptor NKG2A on human tumor-reactive CD8 T cells Spotlight 

Fesneau et al. showed that in HNSCC and CRC tumors, expression of the inhibitor NKG2A, a receptor for HLA-E that is associated with poor prognosis for many solid cancers, was upregulated upon differentiation of CD8+ T cells into cytotoxic tumor-reactive CD39+CD103+ double positive (DP) cells. The NKG2ADP and NKG2A+DP CD8+ T cell TCR repertoires overlapped, suggesting both shared origins and antitumor specificities. NKG2A upregulation on naive CD8+ T cells depended on IL-12 (paradoxically, a promoter of cytotoxic immune responses), TCR and TGFβ stimulation, CD4+ T cells, MHC-II+ cells, and CD40L/CD40 interactions.

Contributed by Paula Hochman

Fesneau et al. showed that in HNSCC and CRC tumors, expression of the inhibitor NKG2A, a receptor for HLA-E that is associated with poor prognosis for many solid cancers, was upregulated upon differentiation of CD8+ T cells into cytotoxic tumor-reactive CD39+CD103+ double positive (DP) cells. The NKG2ADP and NKG2A+DP CD8+ T cell TCR repertoires overlapped, suggesting both shared origins and antitumor specificities. NKG2A upregulation on naive CD8+ T cells depended on IL-12 (paradoxically, a promoter of cytotoxic immune responses), TCR and TGFβ stimulation, CD4+ T cells, MHC-II+ cells, and CD40L/CD40 interactions.

Contributed by Paula Hochman

ABSTRACT: Blockade of NKG2A/HLA-E interaction is a promising strategy to unleash the anti-tumor response. Yet the role of NKG2A(+) CD8 T cells in the anti-tumor response and the regulation of NKG2A expression on human tumor-infiltrating T cells are still poorly understood. Here, by performing CITE-seq on T cells derived from head and neck squamous cell carcinoma and colorectal cancer, we show that NKG2A expression is induced on CD8 T cells differentiating into cytotoxic, CD39(+)CD103(+) double positive (DP) cells, a phenotype associated with tumor-reactive T cells. This developmental trajectory leads to TCR repertoire overlap between the NKG2A(-) and NKG2A(+) DP CD8 T cells, suggesting shared antigen specificities. Mechanistically, IL-12 is essential for the expression of NKG2A on CD8 T cells in a CD40/CD40L- dependent manner, in conjunction with TCR stimulation. Our study thus reveals that NKG2A is induced by IL-12 on human tumor-reactive CD8 T cells exposed to a TGF-_-rich environment, highlighting an underappreciated immuno-regulatory feedback loop dependent on IL-12 stimulation.

Author Info: (1) Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA. (2) Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA. (3)

Author Info: (1) Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA. (2) Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA. (3) Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA. (4) Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA. (5) Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA. (6) Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA. (7) Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA. (8) Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA. thomas.duhen@providence.org.

Platelet factor 4-induced TH1-Treg polarization suppresses antitumor immunity

Spotlight 

Kuratani et al. demonstrated that VeDTR-mediated selective deletion of arginase I (Arg1)-expressing tumor-associated macrophages (Arg1+ TAMs) reduced the ratio of TH1-Treg cells in the TME, and inhibited tumor growth in subcutaneously implanted MC38 and B16F10 tumors. Arg1+ TAMs secreted the chemokine platelet factor 4 (PF4 [Cxcl4]), which polarized Tregs into TH1-Tregs in a CXCR3-dependent manner. Both genetic PF4 inactivation and mAb-mediated PF4 neutralization enhanced antitumor immunity and reduced tumor growth and the TH1-Treg ratio in the TME.

Contributed by Shishir Pant

Kuratani et al. demonstrated that VeDTR-mediated selective deletion of arginase I (Arg1)-expressing tumor-associated macrophages (Arg1+ TAMs) reduced the ratio of TH1-Treg cells in the TME, and inhibited tumor growth in subcutaneously implanted MC38 and B16F10 tumors. Arg1+ TAMs secreted the chemokine platelet factor 4 (PF4 [Cxcl4]), which polarized Tregs into TH1-Tregs in a CXCR3-dependent manner. Both genetic PF4 inactivation and mAb-mediated PF4 neutralization enhanced antitumor immunity and reduced tumor growth and the TH1-Treg ratio in the TME.

Contributed by Shishir Pant

ABSTRACT: The tumor microenvironment (TME) contains a number of immune-suppressive cells such as T helper 1-polarized regulatory T cells (T(H)1-T(reg) cells). However, little is known about the mechanism behind the abundant presence of T(H)1-T(reg) cells in the TME. We demonstrate that selective depletion of arginase I (Arg1)-expressing tumor-associated macrophages (Arg1(+) TAMs) inhibits tumor growth and concurrently reduces the ratio of T(H)1-T(reg) cells in the TME. Arg1(+) TAMs secrete the chemokine platelet factor 4 (PF4), which reinforces interferon-_ (IFN-_)-induced T(reg) cell polarization into T(H)1-T(reg) cells in a manner dependent on CXCR3 and the IFN-_ receptor. Both genetic PF4 inactivation and PF4 neutralization hinder T(H)1-T(reg) cell accumulation in the TME and reduce tumor growth. Collectively, our study highlights the importance of Arg1(+) TAM-produced PF4 for high T(H)1-T(reg) cell levels in the TME to suppress antitumor immunity.

Author Info: (1) Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan. Laboratory of Immunoparasitology, WPI Immunology Frontier Re

Author Info: (1) Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan. Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan. (2) Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan. Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan. (3) Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan. Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan. (4) Genome Information Research Center, Osaka University, Suita, Osaka, Japan. (5) Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan. Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan. Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan. Center for Advances Modalities and Drug Delivery Systems, Osaka University, Suita, Osaka, Japan. (6) Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan. (7) Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan. Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan. Center for Advances Modalities and Drug Delivery Systems, Osaka University, Suita, Osaka, Japan. Department of Immunochemistry, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan. (8) Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan. Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan. Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan. Center for Advances Modalities and Drug Delivery Systems, Osaka University, Suita, Osaka, Japan.

Ablation of FAS confers allogeneic CD3- CAR T cells with resistance to rejection by T cells and natural killer cells

Spotlight 

Using a genome-wide CRISPR screen, Menegatti et al. identified targets that could extend the persistence of allogeneic C57BL/6 T cells in a BALB/c host. Although disruption of either Fas or B2m improved the survival of allogeneic T cells, Fas KO was more effective than B2m KO in resisting NK cell-mediated allorejection. In human CAR T cells, disruption of TRAC and Fas, compared to TRAC and B2m, improved survival in in vitro and in vivo models of T cell or NK cell-mediated allorejection, and led to superior effector function and tumor control, even without allogeneic pressure.

Contributed by Alex Najibi

Using a genome-wide CRISPR screen, Menegatti et al. identified targets that could extend the persistence of allogeneic C57BL/6 T cells in a BALB/c host. Although disruption of either Fas or B2m improved the survival of allogeneic T cells, Fas KO was more effective than B2m KO in resisting NK cell-mediated allorejection. In human CAR T cells, disruption of TRAC and Fas, compared to TRAC and B2m, improved survival in in vitro and in vivo models of T cell or NK cell-mediated allorejection, and led to superior effector function and tumor control, even without allogeneic pressure.

Contributed by Alex Najibi

ABSTRACT: Allogeneic chimaeric antigen receptor T cells (allo-CAR T cells) derived from healthy donors could provide rapid access to standardized and affordable batches of therapeutic cells if their rejection by the host's immune system is avoided. Here, by means of an in vivo genome-wide CRISPR knockout screen, we show that the deletion of Fas or B2m in allo- T cells increases their survival in immunocompetent mice. Human B2M(-) allo-CAR T cells become highly sensitive to rejection mediated by natural killer (NK) cells, whereas FAS(-) CAR T cells expressing normal levels of human leukocyte antigen I remain resistant to NK cells. CD3(-) FAS(-) CAR T cells outperformed CD3(-) B2M(-) CAR T cells in the control of leukaemia growth in mice under allogeneic pressure by T cells and NK cells. The partial protection of CD3(-) FAS(-) allo-CAR T cells from cellular rejection may improve the efficacy of allogeneic cellular therapies in patients with cancer.

Author Info: (1) Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France. CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France. (2) Im

Author Info: (1) Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France. CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France. (2) Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France. (3) Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France. (4) Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France. CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France. (5) Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France. CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France. (6) Gustave Roussy, Paris-Saclay University, INSERM U1015, Villejuif, France. (7) Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France. (8) Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France. CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France. (9) Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France. (10) Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France. (11) Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France. CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France. (12) Gustave Roussy, Paris-Saclay University, INSERM U1015, Villejuif, France. (13) Gustave Roussy, Paris-Saclay University, INSERM U1015, Villejuif, France. (14) Mnemo Therapeutics, Paris, France. (15) Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA. Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA. Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA. (16) Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA. Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA. Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA. (17) Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA. Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA. Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA. (18) Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France. (19) Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France. sebastian.amigorena@curie.fr. CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France. sebastian.amigorena@curie.fr. Mnemo Therapeutics, Paris, France. sebastian.amigorena@curie.fr. (20) Gustave Roussy, Paris-Saclay University, INSERM U1015, Villejuif, France. Laurie.MENGER@gustaveroussy.fr.

PD-1 and CD73 on naive CD4+ T cells synergistically limit responses to self

Featured  

Nettersheim, Brunel, Sinkovits, et al. confirmed that CD4+ T cells reactive to self antigens expand poorly after vaccination and explored the mechanisms behind this effect. The differences in expansion between CD4+ T cells reactive to self antigens vs. those reactive to foreign antigens could not be explained by differences in the naive repertoires or by an enhanced Treg signature in the self-reactive CD4+ T cells. The self-reactive CD4+ T cells coexpressed CD73 and PD-1, which was found to be responsible for their limited expansion upon vaccination.

Nettersheim, Brunel, Sinkovits, et al. confirmed that CD4+ T cells reactive to self antigens expand poorly after vaccination and explored the mechanisms behind this effect. The differences in expansion between CD4+ T cells reactive to self antigens vs. those reactive to foreign antigens could not be explained by differences in the naive repertoires or by an enhanced Treg signature in the self-reactive CD4+ T cells. The self-reactive CD4+ T cells coexpressed CD73 and PD-1, which was found to be responsible for their limited expansion upon vaccination.

ABSTRACT: Vaccination with self- and foreign peptides induces weak and strong expansion of antigen-specific CD4(+) T cells, respectively, but the mechanism is not known. In the present study, we used computational analysis of the entire mouse major histocompatibility complex class II peptidome to test how much of the naive CD4(+) T cell repertoire specific for self-antigens was shaped by negative selection in the thymus and found that negative selection only partially explained the difference between responses to self and foreign. In naive uninfected and unimmunized mice, we identified higher expression of programmed cell death protein 1 (PD-1) and CD73 mRNA and protein on self-specific CD4(+) T cells compared with foreign-specific CD4(+) T cells. Pharmacological or genetic blockade of PD-1 and CD73 significantly increased the vaccine-induced expansion of self-specific CD4(+) T cells and their transcriptomes were similar to those of foreign-specific CD4(+) T cells. We concluded that PD-1 and CD73 synergistically limited CD4(+) T cell responses to self. These observations have implications for the development of tolerogenic vaccines and cancer immunotherapy.

Author Info: (1) La Jolla Institute for Immunology, La Jolla, CA, USA. (2) La Jolla Institute for Immunology, La Jolla, CA, USA. (3) San Diego Supercomputer Center, University of California, La

Author Info: (1) La Jolla Institute for Immunology, La Jolla, CA, USA. (2) La Jolla Institute for Immunology, La Jolla, CA, USA. (3) San Diego Supercomputer Center, University of California, La Jolla, CA, USA. (4) La Jolla Institute for Immunology, La Jolla, CA, USA. (5) La Jolla Institute for Immunology, La Jolla, CA, USA. Immunology Center of Georgia, Augusta University, Augusta, GA, USA. (6) La Jolla Institute for Immunology, La Jolla, CA, USA. (7) La Jolla Institute for Immunology, La Jolla, CA, USA. Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan. (8) La Jolla Institute for Immunology, La Jolla, CA, USA. Immunology Center of Georgia, Augusta University, Augusta, GA, USA. (9) Immunology Center of Georgia, Augusta University, Augusta, GA, USA. (10) La Jolla Institute for Immunology, La Jolla, CA, USA. (11) Immunology Center of Georgia, Augusta University, Augusta, GA, USA. (12) Immunology Center of Georgia, Augusta University, Augusta, GA, USA. (13) La Jolla Institute for Immunology, La Jolla, CA, USA. (14) La Jolla Institute for Immunology, La Jolla, CA, USA. (15) La Jolla Institute for Immunology, La Jolla, CA, USA. kley@augusta.edu. Immunology Center of Georgia, Augusta University, Augusta, GA, USA. kley@augusta.edu.

IL-2/anti-IL-2 antibody complexes augment immune responses to therapeutic cancer vaccines Spotlight 

An effective scaffold-based vaccine (Vax) promoted massive and preferential monocyte expansion in the LNs. Addition of IL-2/anti-IL-2 (IL-2cx) significantly improved LN cDC1 expansion and activation while also enriching NK cells and CD8+ T cells at the Vax site. Vax+IL-2cx increased the number of peripheral, polyfunctional, antigen-specific CD8+ T cells (with a greater proportion of CD44+CD62L- effectors), and improved MC38 tumor control, dependent completely on cDC1s and partially on NK cells. In B16F10 tumors, Vax+IL-2cx reduced GzmB+ CD8+ T cells compared to Vax alone, yet enhanced NK and cDC1 infiltration, leading to modestly improved tumor control.

Contributed by Morgan Janes

An effective scaffold-based vaccine (Vax) promoted massive and preferential monocyte expansion in the LNs. Addition of IL-2/anti-IL-2 (IL-2cx) significantly improved LN cDC1 expansion and activation while also enriching NK cells and CD8+ T cells at the Vax site. Vax+IL-2cx increased the number of peripheral, polyfunctional, antigen-specific CD8+ T cells (with a greater proportion of CD44+CD62L- effectors), and improved MC38 tumor control, dependent completely on cDC1s and partially on NK cells. In B16F10 tumors, Vax+IL-2cx reduced GzmB+ CD8+ T cells compared to Vax alone, yet enhanced NK and cDC1 infiltration, leading to modestly improved tumor control.

Contributed by Morgan Janes

ABSTRACT: One driver of the high failure rates of clinical trials for therapeutic cancer vaccines is likely the inability to sufficiently engage conventional dendritic cells (cDCs), the antigen-presenting cell (APC) subset that is specialized in priming antitumor T cells. Here, we demonstrate that, relative to vaccination with an injectable mesoporous silica rod (MPS) vaccine alone (Vax), combining MPS vaccines with CD122-biased IL-2/anti-IL-2 antibody complexes (IL-2cx) drives ~3-fold expansion of cDCs at the vaccination sites, vaccine-draining lymph nodes, and spleens of treated mice. Furthermore, relative to Vax alone, Vax+IL-2cx led to a ~3-fold increase in the numbers of CD8(+) T cells and ~15-fold increase in the numbers of NK cells at the vaccination site. Notably, with both the model protein antigen OVA as well as various peptide neoantigens, Vax+IL-2cx induced ~5 to 30-fold greater numbers of circulating antigen-specific CD8(+) T cells relative to Vax alone. We further demonstrate that Vax+IL-2cx leads to significantly improved efficacy in the MC38 colon carcinoma model relative to either monotherapy alone, driving complete regressions in 50% of mice in a cDC-dependent manner. Relative to vaccine alone, Vax+IL-2cx led to comparable numbers of CD8(+) T cells, but markedly greater numbers of NK cells and activated cDCs in the B16F10 melanoma tumor microenvironment post-therapy. Taken together, these findings suggest that the administration of factors that engage both the cDC-CD8(+) T cell and cDC-NK cell axes can boost the potency of therapeutic cancer vaccines.

Author Info: (1) John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138. Wyss Institute for Biologically Inspired Engineering at Harvard University,

Author Info: (1) John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138. Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02215. (2) John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138. Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02215. (3) John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138. Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02215. (4) John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138. Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02215. (5) John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138. Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02215. (6) Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA 94080. (7) John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138. Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02215. (8) Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02215. (9) John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138. Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02215. (10) John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138. Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02215. (11) John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138. Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02215. Harvard Program in Biophysics, Harvard University, Cambridge, MA 02138. (12) John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138. Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02215. (13) Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva CH-1211, Switzerland. Ludwig Institute for Cancer Research, Lausanne CH-1005, Switzerland. Agora Cancer Center, Lausanne CH-1005, Switzerland. (14) Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA 94080. Department of Neurological Surgery, University of California, San Francisco, CA (15) Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA 94080. (16) John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138. Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02215.

Impaired development of memory B cells and antibody responses in humans and mice deficient in PD-1 signaling Spotlight 

Ogishi and Kitaoka et al. demonstrated that the PD-1/PD-L1 axis was essential for optimal humoral immunity through T cell-dependent and B cell-intrinsic mechanisms. Patients with PD-1 or PD-L1 deficiency exhibited diminished memory B cell development and antibody production. PD-1 deficiency disrupted c-Myc expression, reduced somatic hypermutation, and impaired Ig class switching in humans and mice. PD-1 deficiency in CD4+ T cells reduced IL-21 production, leading to impaired B cell responses. B cell-specific PD-1 knockout mice showed impaired memory B cell maturation and antibody repertoire diversity.

Contributed by Shishir Pant

Ogishi and Kitaoka et al. demonstrated that the PD-1/PD-L1 axis was essential for optimal humoral immunity through T cell-dependent and B cell-intrinsic mechanisms. Patients with PD-1 or PD-L1 deficiency exhibited diminished memory B cell development and antibody production. PD-1 deficiency disrupted c-Myc expression, reduced somatic hypermutation, and impaired Ig class switching in humans and mice. PD-1 deficiency in CD4+ T cells reduced IL-21 production, leading to impaired B cell responses. B cell-specific PD-1 knockout mice showed impaired memory B cell maturation and antibody repertoire diversity.

Contributed by Shishir Pant

ABSTRACT: T follicular helper (Tfh) cells abundantly express the immunoreceptor programmed cell death protein 1 (PD-1), and the impact of PD-1 deficiency on antibody (Ab)-mediated immunity in mice is associated with compromised Tfh cell functions. Here, we revisited the role of the PD-1-PD-L1 axis on Ab-mediated immunity. Individuals with inherited PD-1 or PD-L1 deficiency had fewer memory B cells and impaired Ab responses, similar to Pdcd1(-/-) and Cd274(-/-)Pdcd1lg2(-/-) mice. PD-1, PD-L1, or both could be detected on the surface of human naive B cells following in vitro activation. PD-1- or PD-L1-deficient B cells had reduced expression of the transcriptional regulator c-Myc and c-Myc-target genes in vivo, and PD-1 deficiency or neutralization of PD-1 or PD-L1 impeded c-Myc expression and Ab production in human B cells isolated in vitro. Furthermore, B cell-specific deletion of Pdcd1 prevented the physiological accumulation of memory B cells in mice. Thus, PD-1 shapes optimal B cell memory and Ab-mediated immunity through B cell-intrinsic and B cell-extrinsic mechanisms, suggesting that B cell dysregulation contributes to infectious and autoimmune complications following anti-PD-1-PD-L1 immunotherapy.

Author Info: (1) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA; The David Rockefeller Graduate Program, Rock

Author Info: (1) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA; The David Rockefeller Graduate Program, Rockefeller University, New York, NY 10065, USA. Electronic address: oogishi-tky@umin.ac.jp. (2) Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto 606-8303, Japan. (3) Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia. (4) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA. (5) Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto 606-8303, Japan; Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama 230-0045, Japan. (6) Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto 606-8303, Japan. (7) Department of Clinical Immunology and Internal Medicine, Strasbourg University Hospital, INSERM UMR-S1109, 67000 Strasbourg, France. (8) Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia. (9) Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia. (10) Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia. (11) Department of Human Immunology, Research Branch, Sidra Medicine, Doha, Qatar. (12) Center for Interventional Immunology, Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, WA 98101, USA. (13) Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia. (14) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA. (15) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA; Primary Immunodeficiencies Group, University of Antioquia UdeA, Medellin, Colombia; School of Microbiology, University of Antioquia UdeA, Medellin, Colombia. (16) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA. (17) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA. (18) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA. (19) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA. (20) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA. (21) Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris CitŽ University, Imagine Institute, Paris, France. (22) Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris CitŽ University, Imagine Institute, Paris, France. (23) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA. (24) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA. (25) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA. (26) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA. (27) Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris CitŽ University, Imagine Institute, Paris, France. (28) Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris CitŽ University, Imagine Institute, Paris, France. (29) McGill University Genome Center, Montreal, QC, Canada; McGill Research Centre on Complex Traits, Dahdaleh Institute of Genomic Medicine, Montreal, QC H3A 0G1, Canada; Department of Human Genetics, McGill University, Montreal, QC H3A 0G1, Canada. (30) Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA. (31) Department of Human Immunology, Research Branch, Sidra Medicine, Doha, Qatar. (32) Department of Human Immunology, Research Branch, Sidra Medicine, Doha, Qatar. (33) Department of Pediatric Immunology, Dr. Sami Ulus Maternity and Children's Health and Diseases Training and Research Hospital, Ankara, Turkey. (34) Center for Systems Immunology, Benaroya Research Institute, Seattle, WA 98101, USA. (35) Center for Systems Immunology, Benaroya Research Institute, Seattle, WA 98101, USA. (36) Department of Immunobiology, School of Immunobiology & Microbial Sciences, Kings' College London, London WC2R 2LS, UK. (37) Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan. (38) Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA. (39) Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA. (40) Department of Immunology and Infectious Diseases, Sydney Children's Hospital, High Street, Randwick, NSW 2031, Australia; School of Women's and Children's Health, University of New South Wales, Sydney, NSW 2052, Australia; Clinical Immunogenomics Research Consortium Australasia (CIRCA), Darlinghurst, NSW 2010, Australia. (41) Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Clinical Immunogenomics Research Consortium Australasia (CIRCA), Darlinghurst, NSW 2010, Australia; Westmead Clinical School, The University of Sydney, Westmead, NSW 2145, Australia. (42) John Hunter Children's Hospital, Newcastle, NSW 2305, Australia. (43) Canterbury Health Laboratories, Christchurch 8140, New Zealand. (44) Department of Pediatrics, Hiroshima University Hospital, Hiroshima 734-0037, Japan. (45) Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-0016, Japan. (46) Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan. (47) Department of Immunobiology, School of Immunobiology & Microbial Sciences, Kings' College London, London WC2R 2LS, UK. (48) Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA. (49) McGill Research Centre on Complex Traits, Dahdaleh Institute of Genomic Medicine, Montreal, QC H3A 0G1, Canada; Department of Biochemistry, McGill University, Montreal, QC H3A 0G1, Canada. (50) Department of Human Immunology, Research Branch, Sidra Medicine, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar. (51) Center for Interventional Immunology, Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, WA 98101, USA; Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, WA 98101, USA. (52) Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter EX1 2ED, UK. (53) Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris CitŽ University, Imagine Institute, Paris, France. (54) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris CitŽ University, Imagine Institute, Paris, France; Center for the Study of Primary Immunodeficiencies, Necker Hospital for Sick Children Assistance Publique-H™pitaux de Paris (AP-HP), Paris, France. (55) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris CitŽ University, Imagine Institute, Paris, France. (56) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris CitŽ University, Imagine Institute, Paris, France. (57) Department of Clinical Immunology and Internal Medicine, Strasbourg University Hospital, INSERM UMR-S1109, 67000 Strasbourg, France. (58) Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia; Clinical Immunogenomics Research Consortium Australasia (CIRCA), Darlinghurst, NSW 2010, Australia. (59) Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter EX1 2ED, UK. (60) Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto 606-8303, Japan; Department of Immuno-Oncology PDT, Kyoto University Graduate School of Medicine, Kyoto 606-8303, Japan. (61) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris CitŽ University, Imagine Institute, Paris, France. (62) Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto 606-8303, Japan. (63) St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris CitŽ University, Imagine Institute, Paris, France; Department of Pediatrics, Necker Hospital for Sick Children, 75015 Paris, France; Howard Hughes Medical Institute, New York, NY 10065, USA. (64) Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia; Clinical Immunogenomics Research Consortium Australasia (CIRCA), Darlinghurst, NSW 2010, Australia. Electronic address: s.tangye@garvan.org.au.

Analysis of PD1, LAG3, TIGIT, and TIM3 expression in human lung adenocarcinoma reveals a 25-gene signature predicting immunotherapy response Spotlight 

Guégan et al. analyzed tumor samples from 166 lung adenocarcinoma patients treated with immune checkpoint inhibitors (ICI) to evaluate the predictive value of CD8+ T cell exhaustion on responsiveness to immunotherapy. Using multiplex immunofluorescence, CD8+ TILs co-expressing high levels of checkpoint molecules PD-1, LAG3, TIGIT, or TIM3 were associated with ICI resistance, irrespective of PD-L1 levels. In addition, a 25-gene CD8+ T cell exhaustion signature was identified that predicted patients’ responses to ICI (but not to TKI) therapy, which was further validated in additional external datasets, including melanoma and renal cell cancer.

Contributed by Katherine Turner

Guégan et al. analyzed tumor samples from 166 lung adenocarcinoma patients treated with immune checkpoint inhibitors (ICI) to evaluate the predictive value of CD8+ T cell exhaustion on responsiveness to immunotherapy. Using multiplex immunofluorescence, CD8+ TILs co-expressing high levels of checkpoint molecules PD-1, LAG3, TIGIT, or TIM3 were associated with ICI resistance, irrespective of PD-L1 levels. In addition, a 25-gene CD8+ T cell exhaustion signature was identified that predicted patients’ responses to ICI (but not to TKI) therapy, which was further validated in additional external datasets, including melanoma and renal cell cancer.

Contributed by Katherine Turner

ABSTRACT: Immune checkpoint inhibitors (ICIs) have advanced the treatment of non-small cell lung cancer (NSCLC). This study evaluates the predictive value of CD8(+) T cell exhaustion in patients with lung adenocarcinoma treated with ICIs. By analyzing tumor samples from 166 patients through multiplex immunofluorescence, we quantify tumor-infiltrating lymphocytes (TILs) expressing exhaustion markers programmed cell death-1 (PD1), lymphocyte activation gene 3 (LAG3), T cell immunoreceptor with Ig and ITIM domains (TIGIT), and T cell immunoglobulin and mucin domain 3 (TIM3). Their co-expression is associated with ICI resistance, irrespective of programmed cell death ligand-1 (PD-L1) status. We also identify a 25-gene signature indicative of CD8(+) T cell exhaustion with high predictive accuracy for ICI response. Validated using several datasets from various clinical trials, this signature accurately predicts ICI responsiveness. Our findings highlight T cell exhaustion's significance in lung adenocarcinoma responses to ICIs and suggest the 25-gene signature as a potential universal biomarker to reinforce precision medicine. This was registered under Clinical Trial registration number NCT02534649.

Author Info: (1) Explicyte Immuno-Oncology, Bordeaux, France. (2) Explicyte Immuno-Oncology, Bordeaux, France; Department of Medicine, Institut Bergonié, Bordeaux, France. (3) Department of Pat

Author Info: (1) Explicyte Immuno-Oncology, Bordeaux, France. (2) Explicyte Immuno-Oncology, Bordeaux, France; Department of Medicine, Institut Bergonié, Bordeaux, France. (3) Department of Pathology, University Hospital Centre of Nice, Nice, France. (4) Explicyte Immuno-Oncology, Bordeaux, France; Department of Medicine, Institut Bergonié, Bordeaux, France. (5) Explicyte Immuno-Oncology, Bordeaux, France; Department of Medicine, Institut Bergonié, Bordeaux, France. (6) Department of Medicine, Institut Bergonié, Bordeaux, France. (7) Department of Medicine, Institut Bergonié, Bordeaux, France. (8) Department of Medicine, Institut Bergonié, Bordeaux, France. (9) Department of Medicine, Institut Bergonié, Bordeaux, France. (10) Department of Medicine, Institut Bergonié, Bordeaux, France. (11) Department of Medicine, Institut Bergonié, Bordeaux, France. (12) Explicyte Immuno-Oncology, Bordeaux, France. (13) Explicyte Immuno-Oncology, Bordeaux, France. (14) Explicyte Immuno-Oncology, Bordeaux, France. (15) Department of Medicine, Institut Bergonié, Bordeaux, France. (16) Department of Pathology, Institut Bergonié, Bordeaux, France. (17) AstraZeneca, Rahway, NJ, USA. (18) AstraZeneca, Rahway, NJ, USA. (19) University of Copenhagen, Copenhagen, Denmark. (20) Clinique Marzet, Pau, France. (21) Centre Hospitalier de la Côte Basque, Bayonne, France. (22) Explicyte Immuno-Oncology, Bordeaux, France. (23) Department of Medicine, Institut Bergonié, Bordeaux, France. Electronic address: a.italiano@bordeaux.unicancer.fr.

PD-1 blockade plus cisplatin-based chemotherapy in patients with small cell/neuroendocrine bladder and prostate cancers Spotlight 

In a phase 1b study, 15 patients with late-stage small cell bladder cancer or small cell/ neuroendocrine prostate cancer were treated with standard-of-care platinum chemotherapy and anti-PD-1 (pembrolizumab). At 24 months, PFS and OS were 50% and 71%, respectively. Treatment was generally safe, with grade 3+ adverse events in 40% of patients. Treatment led to expansion of pre-existing effector, activated, and cytotoxic T cell clonotypes, loss of naive, memory, and regulatory T cells, and detection of majority-novel clonotypes. T cell clonal expansion, but not baseline PD-L1 expression, TMB, or MSI status, correlated with PFS.

Contributed by Alex Najibi

In a phase 1b study, 15 patients with late-stage small cell bladder cancer or small cell/ neuroendocrine prostate cancer were treated with standard-of-care platinum chemotherapy and anti-PD-1 (pembrolizumab). At 24 months, PFS and OS were 50% and 71%, respectively. Treatment was generally safe, with grade 3+ adverse events in 40% of patients. Treatment led to expansion of pre-existing effector, activated, and cytotoxic T cell clonotypes, loss of naive, memory, and regulatory T cells, and detection of majority-novel clonotypes. T cell clonal expansion, but not baseline PD-L1 expression, TMB, or MSI status, correlated with PFS.

Contributed by Alex Najibi

ABSTRACT: Small cell neuroendocrine cancers share biologic similarities across tissue types, including transient response to platinum-based chemotherapy with rapid progression of disease. We report a phase 1b study of pembrolizumab in combination with platinum-based chemotherapy in 15 patients with stage III-IV small cell bladder (cohort 1) or small cell/neuroendocrine prostate cancers (cohort 2). Overall response rate (ORR) is 43% with two-year overall survival (OS) rate of 86% (95% confidence interval [CI]: 0.63, 1.00) for cohort 1 and 57% (95% CI: 0.30, 1.00) for cohort 2. Treatment is tolerated well with grade 3 or higher adverse events occurring in 40% of patients with no deaths or treatment cessation secondary to toxicity. Single-cell and T cell receptor sequencing of serial peripheral blood samples reveals clonal expansion of diverse T cell repertoire correlating with progression-free survival. Our results demonstrate promising efficacy and safety of this treatment combination and support future investigation of this biomarker. This study was registered at ClinicalTrials.gov (NCT03582475).

Author Info: (1) Department of Molecular, Cellular and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA. (2) Department of Urology, David Geffen School of Medi

Author Info: (1) Department of Molecular, Cellular and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA. (2) Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA. (3) Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA. (4) Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA. (5) Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA. (6) Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA. (7) Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA. (8) Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA. (9) Department of Molecular, Cellular and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA. (10) Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA. (11) Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. (12) Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. (13) Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; The VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA. (14) Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA. Electronic address: arnoldchin@mednet.ucla.edu.

A tetraspecific engager armed with a non-alpha IL-2 variant harnesses natural killer cells against B cell non-Hodgkin lymphoma Featured  

To capitalize on the antitumor potential of NK cells for the treatment of B cell non-Hodgkin lymphoma, Demaria et al. evaluated a tetraspecific NK cell engager (NKCE) targeting NKp46 and CD16a on NK cells and CD20 on tumor cells, and armed with a non-alpha IL-2 variant (CD20-NKCE-IL2). In a series of in vitro and in vivo studies, CD20-NKCE-IL2v boosted NK cell proliferation, cytotoxicity, activating receptors, and tumor homing, ultimately supporting antitumor activity, even against tumor cells with low or no CD20 and tumor cells expressing MHC-I. Compared to similar treatments, including T cell engagers, CD20-NKCE-IL2 showed stronger antitumor efficacy and reduced toxicity.

To capitalize on the antitumor potential of NK cells for the treatment of B cell non-Hodgkin lymphoma, Demaria et al. evaluated a tetraspecific NK cell engager (NKCE) targeting NKp46 and CD16a on NK cells and CD20 on tumor cells, and armed with a non-alpha IL-2 variant (CD20-NKCE-IL2). In a series of in vitro and in vivo studies, CD20-NKCE-IL2v boosted NK cell proliferation, cytotoxicity, activating receptors, and tumor homing, ultimately supporting antitumor activity, even against tumor cells with low or no CD20 and tumor cells expressing MHC-I. Compared to similar treatments, including T cell engagers, CD20-NKCE-IL2 showed stronger antitumor efficacy and reduced toxicity.

ABSTRACT: NK cells offer a promising alternative to T cell therapies in cancer. We evaluated IPH6501, a clinical-stage, tetraspecific NK cell engager (NKCE) armed with a non-alpha IL-2 variant (IL-2v), which targets CD20 and was developed for treating B cell non-Hodgkin lymphoma (B-NHL). CD20-NKCE-IL2v boosts NK cell proliferation and cytotoxicity, showing activity against a range of B-NHL cell lines, including those with low CD20 density. Whereas it presented reduced toxicities compared with those commonly associated with T cell therapies, CD20-NKCE-IL2v showed greater killing efficacy over a T cell engager targeting CD20 in in vitro preclinical models. CD20-NKCE-IL2v also increased the cell surface expression of NK cell-activating receptors, leading to activity against CD20-negative tumor cells. In vivo studies in nonhuman primates and tumor mouse models further validated its efficacy and revealed that CD20-NKCE-IL2v induces peripheral NK cell homing at the tumor site. CD20-NKCE-IL2v emerges as a potential alternative in the treatment landscape of B-NHL.

Author Info: (1) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (2) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (3) Innate Pharma Research Lab

Author Info: (1) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (2) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (3) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (4) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (5) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (6) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (7) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (8) Aix Marseille UniversitŽ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France. (9) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (10) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (11) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (12) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (13) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (14) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (15) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (16) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (17) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (18) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (19) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (20) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (21) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (22) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (23) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (24) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (25) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (26) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (27) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (28) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (29) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (30) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (31) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (32) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (33) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. (34) Innate Pharma Research Laboratories, Innate Pharma, Marseille, France. Aix Marseille UniversitŽ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France. APHM, H™pital de la Timone, Marseille-Immunop™le Profiling Platform, Marseille, France. Paris-Saclay Cancer Cluster, Le Kremlin-Bictre, France. UniversitŽ Paris-Saclay, Gustave Roussy, INSERM, PrŽdicteurs molŽculaires et nouvelles cibles en oncologie, 94800, Villejuif, France.

A bispecific CD40 agonistic antibody allowing for antibody-peptide conjugate formation to enable cancer-specific peptide delivery, resulting in improved T proliferation and anti-tumor immunity in mice Spotlight 

Mebrahtu et al. fused a CD40 agonist mAb with an scFv that binds a universal peptide tag handle (pTag), enabling modular loading of pTag-containing antigen peptides, which promoted antigen-specific T cell responses when bound to vaccine peptides. The final BiAb, containing an IgG2 CD40 agonist mAb fused to the scFv/pTag via the heavy chain, was chosen for its optimal yield and agonist activity. The BiAb elicited significant antigen-specific T cell proliferation in the LNs and spleen, which correlated inversely with pTag length. In the TC1 and MC38 models, the BiAb improved tumor control compared to CD40 agonist + free peptide or agonist + CpG.

Contributed by Morgan Janes

Mebrahtu et al. fused a CD40 agonist mAb with an scFv that binds a universal peptide tag handle (pTag), enabling modular loading of pTag-containing antigen peptides, which promoted antigen-specific T cell responses when bound to vaccine peptides. The final BiAb, containing an IgG2 CD40 agonist mAb fused to the scFv/pTag via the heavy chain, was chosen for its optimal yield and agonist activity. The BiAb elicited significant antigen-specific T cell proliferation in the LNs and spleen, which correlated inversely with pTag length. In the TC1 and MC38 models, the BiAb improved tumor control compared to CD40 agonist + free peptide or agonist + CpG.

Contributed by Morgan Janes

ABSTRACT: Current antibody-based immunotherapy depends on tumor antigen shedding for proper T cell priming. Here we select a novel human CD40 agonistic drug candidate and generate a bispecific antibody, herein named BiA9*2_HF, that allows for rapid antibody-peptide conjugate formation. The format is designed to facilitate peptide antigen delivery to CD40 expressing cells combined with simultaneous CD40 agonistic activity. In vivo, the selected bispecific antibody BiA9*2_HF loaded with peptide cargos induces improved antigen-specific proliferation of CD8+ (10-15 fold) and CD4+ T cells (2-7 fold) over control in draining lymph nodes. In both virus-induced and neoantigen-based mouse tumor models, BiA9*2_HF demonstrates therapeutic efficacy and elevated safety profile, with complete tumor clearance, as well as measured abscopal impact on tumor growth. The BiA9*2_HF drug candidate can thus be utilized to tailor immunotherapeutics for cancer patients.

Author Info: (1) KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden. Strike Pharma AB, Up

Author Info: (1) KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden. Strike Pharma AB, Uppsala, Sweden. (2) Strike Pharma AB, Uppsala, Sweden. Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, Sweden. (3) Strike Pharma AB, Uppsala, Sweden. (4) Strike Pharma AB, Uppsala, Sweden. (5) Strike Pharma AB, Uppsala, Sweden. Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, Sweden. (6) Strike Pharma AB, Uppsala, Sweden. Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, Sweden. (7) Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden. (8) KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden. Science for Life Laboratory, Drug Discovery and Development, Stockholm, Sweden. (9) KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden. Science for Life Laboratory, Drug Discovery and Development, Stockholm, Sweden. (10) KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden. Science for Life Laboratory, Drug Discovery and Development, Stockholm, Sweden. (11) KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden. Science for Life Laboratory, Drug Discovery and Development, Stockholm, Sweden. (12) KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden. Science for Life Laboratory, Drug Discovery and Development, Stockholm, Sweden. (13) Strike Pharma AB, Uppsala, Sweden. SciCross AB, Skšvde, Sweden. (14) KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden. johanr@biotech.kth.se. Strike Pharma AB, Uppsala, Sweden. johanr@biotech.kth.se. (15) Strike Pharma AB, Uppsala, Sweden. sara.mangsbo@farmaci.uu.se. Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, Sweden. sara.mangsbo@farmaci.uu.se.

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.