Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Cancer Cell ; 42(4): 623-645.e10, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38490212

ABSTRACT

Genes limiting T cell antitumor activity may serve as therapeutic targets. It has not been systematically studied whether there are regulators that uniquely or broadly contribute to T cell fitness. We perform genome-scale CRISPR-Cas9 knockout screens in primary CD8 T cells to uncover genes negatively impacting fitness upon three modes of stimulation: (1) intense, triggering activation-induced cell death (AICD); (2) acute, triggering expansion; (3) chronic, causing dysfunction. Besides established regulators, we uncover genes controlling T cell fitness either specifically or commonly upon differential stimulation. Dap5 ablation, ranking highly in all three screens, increases translation while enhancing tumor killing. Loss of Icam1-mediated homotypic T cell clustering amplifies cell expansion and effector functions after both acute and intense stimulation. Lastly, Ctbp1 inactivation induces functional T cell persistence exclusively upon chronic stimulation. Our results functionally annotate fitness regulators based on their unique or shared contribution to traits limiting T cell antitumor activity.


Subject(s)
CRISPR-Cas Systems , Neoplasms , Humans , CD8-Positive T-Lymphocytes , Neoplasms/genetics
2.
Front Immunol ; 14: 1162706, 2023.
Article in English | MEDLINE | ID: mdl-37398651

ABSTRACT

Functional genetic screens to uncover tumor-intrinsic nodes of immune resistance have uncovered numerous mechanisms by which tumors evade our immune system. However, due to technical limitations, tumor heterogeneity is imperfectly captured with many of these analyses. Here, we provide an overview of the nature and sources of heterogeneity that are relevant for tumor-immune interactions. We argue that this heterogeneity may actually contribute to the discovery of novel mechanisms of immune evasion, given a sufficiently large and heterogeneous set of input data. Taking advantage of tumor cell heterogeneity, we provide proof-of-concept analyses of mechanisms of TNF resistance. Thus, consideration of tumor heterogeneity is imperative to increase our understanding of immune resistance mechanisms.


Subject(s)
CRISPR-Cas Systems , Neoplasms , Humans , Neoplasms/genetics
4.
Cell Rep Med ; 4(2): 100941, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36812891

ABSTRACT

By restoring tryptophan, indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors aim to reactivate anti-tumor T cells. However, a phase III trial assessing their clinical benefit failed, prompting us to revisit the role of IDO1 in tumor cells under T cell attack. We show here that IDO1 inhibition leads to an adverse protection of melanoma cells to T cell-derived interferon-gamma (IFNγ). RNA sequencing and ribosome profiling shows that IFNγ shuts down general protein translation, which is reversed by IDO1 inhibition. Impaired translation is accompanied by an amino acid deprivation-dependent stress response driving activating transcription factor-4 (ATF4)high/microphtalmia-associated transcription factor (MITF)low transcriptomic signatures, also in patient melanomas. Single-cell sequencing analysis reveals that MITF downregulation upon immune checkpoint blockade treatment predicts improved patient outcome. Conversely, MITF restoration in cultured melanoma cells causes T cell resistance. These results highlight the critical role of tryptophan and MITF in the melanoma response to T cell-derived IFNγ and uncover an unexpected negative consequence of IDO1 inhibition.


Subject(s)
Melanoma , Tryptophan , Humans , Melanoma/pathology , Interferon-gamma/metabolism , T-Lymphocytes/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
5.
EMBO J ; 42(5): e111614, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36715448

ABSTRACT

Resistance to cancer immunotherapy continues to impair common clinical benefit. Here, we use whole-genome CRISPR-Cas9 knockout data to uncover an important role for Tuberous Sclerosis Complex 2 (TSC2) in determining tumor susceptibility to cytotoxic T lymphocyte (CTL) killing in human melanoma cells. TSC2-depleted tumor cells had disrupted mTOR regulation following CTL attack, which was associated with enhanced cell death. Wild-type tumor cells adapted to CTL attack by shifting their mTOR signaling balance toward increased mTORC2 activity, circumventing apoptosis, and necroptosis. TSC2 ablation strongly augmented tumor cell sensitivity to CTL attack in vitro and in vivo, suggesting one of its functions is to critically protect tumor cells. Mechanistically, TSC2 inactivation caused elevation of TRAIL receptor expression, cooperating with mTORC1-S6 signaling to induce tumor cell death. Clinically, we found a negative correlation between TSC2 expression and TRAIL signaling in TCGA patient cohorts. Moreover, a lower TSC2 immune response signature was observed in melanomas from patients responding to immune checkpoint blockade. Our study uncovers a pivotal role for TSC2 in the cancer immune response by governing crosstalk between TSC2-mTOR and TRAIL signaling, aiding future therapeutic exploration of this pathway in immuno-oncology.


Subject(s)
Tuberous Sclerosis , Tumor Suppressor Proteins , Humans , Cell Death , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 2 Protein/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
Cell Rep Med ; 3(6): 100655, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35688159

ABSTRACT

Tumor escape mechanisms for immunotherapy include deficiencies in antigen presentation, diminishing adaptive CD8+ T cell antitumor activity. Although innate natural killer (NK) cells are triggered by loss of MHC class I, their response is often inadequate. To increase tumor susceptibility to both innate and adaptive immune elimination, we performed parallel genome-wide CRISPR-Cas9 knockout screens under NK and CD8+ T cell pressure. We identify all components, RNF31, RBCK1, and SHARPIN, of the linear ubiquitination chain assembly complex (LUBAC). Genetic and pharmacologic ablation of RNF31, an E3 ubiquitin ligase, strongly sensitizes cancer cells to NK and CD8+ T cell killing. This occurs in a tumor necrosis factor (TNF)-dependent manner, causing loss of A20 and non-canonical IKK complexes from TNF receptor complex I. A small-molecule RNF31 inhibitor sensitizes colon carcinoma organoids to TNF and greatly enhances bystander killing of MHC antigen-deficient tumor cells. These results merit exploration of RNF31 inhibition as a clinical pharmacological opportunity for immunotherapy-refractory cancers.


Subject(s)
Tumor Escape , Ubiquitin-Protein Ligases , Killer Cells, Natural , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
7.
Nat Commun ; 13(1): 1923, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35395848

ABSTRACT

The cytokine IFNγ differentially impacts on tumors upon immune checkpoint blockade (ICB). Despite our understanding of downstream signaling events, less is known about regulation of its receptor (IFNγ-R1). With an unbiased genome-wide CRISPR/Cas9 screen for critical regulators of IFNγ-R1 cell surface abundance, we identify STUB1 as an E3 ubiquitin ligase for IFNγ-R1 in complex with its signal-relaying kinase JAK1. STUB1 mediates ubiquitination-dependent proteasomal degradation of IFNγ-R1/JAK1 complex through IFNγ-R1K285 and JAK1K249. Conversely, STUB1 inactivation amplifies IFNγ signaling, sensitizing tumor cells to cytotoxic T cells in vitro. This is corroborated by an anticorrelation between STUB1 expression and IFNγ response in ICB-treated patients. Consistent with the context-dependent effects of IFNγ in vivo, anti-PD-1 response is increased in heterogenous tumors comprising both wildtype and STUB1-deficient cells, but not full STUB1 knockout tumors. These results uncover STUB1 as a critical regulator of IFNγ-R1, and highlight the context-dependency of STUB1-regulated IFNγ signaling for ICB outcome.


Subject(s)
Interferon-gamma , Neoplasms , Receptors, Interferon , Ubiquitin-Protein Ligases , Humans , Immune Checkpoint Inhibitors , Interferon-gamma/metabolism , Neoplasms/immunology , Receptors, Interferon/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Interferon gamma Receptor
8.
Science ; 373(6559): 1088-1089, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34516853

ABSTRACT

Melanoma can arise only from cells with a permissive chromatin landscape.


Subject(s)
Oncogenes
9.
Clin Cancer Res ; 27(19): 5389-5400, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34230026

ABSTRACT

PURPOSE: Combining anti-PD-1 + anti-CTLA-4 immune-checkpoint blockade (ICB) shows improved patient benefit, but it is associated with severe immune-related adverse events and exceedingly high cost. Therefore, there is a dire need to predict which patients respond to monotherapy and which require combination ICB treatment. EXPERIMENTAL DESIGN: In patient-derived melanoma xenografts (PDX), human tumor microenvironment (TME) cells were swiftly replaced by murine cells upon transplantation. Using our XenofilteR deconvolution algorithm we curated human tumor cell RNA reads, which were subsequently subtracted in silico from bulk (tumor cell + TME) patients' melanoma RNA. This produced a purely tumor cell-intrinsic signature ("InTumor") and a signature comprising tumor cell-extrinsic RNA reads ("ExTumor"). RESULTS: We show that whereas the InTumor signature predicts response to anti-PD-1, the ExTumor predicts anti-CTLA-4 benefit. In PDX, InTumorLO, but not InTumorHI, tumors are effectively eliminated by cytotoxic T cells. When used in conjunction, the InTumor and ExTumor signatures identify not only patients who have a substantially higher chance of responding to combination treatment than to either monotherapy, but also those who are likely to benefit little from anti-CTLA-4 on top of anti-PD-1. CONCLUSIONS: These signatures may be exploited to distinguish melanoma patients who need combination ICB blockade from those who likely benefit from either monotherapy.


Subject(s)
Melanoma , Programmed Cell Death 1 Receptor , Animals , CTLA-4 Antigen , Humans , Immune Checkpoint Inhibitors , Melanoma/drug therapy , Melanoma/genetics , Mice , Programmed Cell Death 1 Receptor/therapeutic use , RNA , Tumor Microenvironment
10.
Cancer Res ; 81(7): 1775-1787, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33531370

ABSTRACT

Although immune checkpoint blockade (ICB) has shown remarkable clinical benefit in a subset of patients with melanoma and lung cancer, most patients experience no durable benefit. The receptor tyrosine kinase AXL is commonly implicated in therapy resistance and may serve as a marker for therapy-refractory tumors, for example in melanoma, as we previously demonstrated. Here, we show that enapotamab vedotin (EnaV), an antibody-drug conjugate targeting AXL, effectively targets tumors that display insensitivity to immunotherapy or tumor-specific T cells in several melanoma and lung cancer models. In addition to its direct tumor cell killing activity, EnaV treatment induced an inflammatory response and immunogenic cell death in tumor cells and promoted the induction of a memory-like phenotype in cytotoxic T cells. Combining EnaV with tumor-specific T cells proved superior to either treatment alone in models of melanoma and lung cancer and induced ICB benefit in models otherwise insensitive to anti-PD-1 treatment. Our findings indicate that targeting AXL-expressing, immunotherapy-resistant tumors with EnaV causes an immune-stimulating tumor microenvironment and enhances sensitivity to ICB, warranting further investigation of this treatment combination. SIGNIFICANCE: These findings show that targeting AXL-positive tumor fractions with an antibody-drug conjugate enhances antitumor immunity in several humanized tumor models of melanoma and lung cancer.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Immunoconjugates/therapeutic use , Lung Neoplasms/therapy , Melanoma/therapy , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Combined Modality Therapy , Drug Resistance, Neoplasm/immunology , Drug Synergism , HEK293 Cells , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immunoconjugates/administration & dosage , Immunotherapy , Lung Neoplasms/pathology , Male , Melanoma/pathology , Mice , Mice, Nude , Mice, Transgenic , Molecular Targeted Therapy/methods , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
11.
Nat Protoc ; 16(2): 791-811, 2021 02.
Article in English | MEDLINE | ID: mdl-33349704

ABSTRACT

Human skin harbors various immune cells that are crucial for the control of injury and infection. However, the current understanding of immune cell function within viable human skin tissue is limited. We developed an ex vivo imaging approach in which fresh skin biopsies are mounted and then labeled with nanobodies or antibodies against cell surface markers on tissue-resident memory CD8+ T cells, other immune cells of interest, or extracellular tissue components. Subsequent longitudinal imaging allows one to describe the dynamic behavior of human skin-resident cells in situ. In addition, this strategy can be used to study immune cell function in murine skin. The ability to follow the spatiotemporal behavior of CD8+ T cells and other immune cells in skin, including their response to immune stimuli, provides a platform to investigate physiological immune cell behavior and immune cell behavior in skin diseases. The mounting, staining and imaging of skin samples requires ~1.5 d, and subsequent tracking analysis requires a minimum of 1 d. The optional production of fluorescently labeled nanobodies takes ~5 d.


Subject(s)
Skin/immunology , Skin/pathology , Staining and Labeling/methods , Animals , Biopsy/methods , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Culture Techniques/methods , Humans , Mice , Skin/cytology
12.
Nat Commun ; 11(1): 3946, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32770055

ABSTRACT

Melanomas can switch to a dedifferentiated cell state upon exposure to cytotoxic T cells. However, it is unclear whether such tumor cells pre-exist in patients and whether they can be resensitized to immunotherapy. Here, we chronically expose (patient-derived) melanoma cell lines to differentiation antigen-specific cytotoxic T cells and observe strong enrichment of a pre-existing NGFRhi population. These fractions are refractory also to T cells recognizing non-differentiation antigens, as well as to BRAF + MEK inhibitors. NGFRhi cells induce the neurotrophic factor BDNF, which contributes to T cell resistance, as does NGFR. In melanoma patients, a tumor-intrinsic NGFR signature predicts anti-PD-1 therapy resistance, and NGFRhi tumor fractions are associated with immune exclusion. Lastly, pharmacologic NGFR inhibition restores tumor sensitivity to T cell attack in vitro and in melanoma xenografts. These findings demonstrate the existence of a stable and pre-existing NGFRhi multitherapy-refractory melanoma subpopulation, which ought to be eliminated to revert intrinsic resistance to immunotherapeutic intervention.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Melanoma/drug therapy , Nerve Tissue Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, Nerve Growth Factor/metabolism , Skin Neoplasms/drug therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Antineoplastic Agents, Immunological/therapeutic use , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Male , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , RNA-Seq , Receptors, Nerve Growth Factor/antagonists & inhibitors , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Escape/genetics , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
13.
J Exp Med ; 217(10)2020 10 05.
Article in English | MEDLINE | ID: mdl-32728699

ABSTRACT

An increasing body of evidence emphasizes the role of tissue-resident memory T cells (TRM) in the defense against recurring pathogens and malignant neoplasms. However, little is known with regard to the origin of these cells and their kinship to other CD8+ T cell compartments. To address this issue, we followed the antigen-specific progeny of individual naive CD8+ T cells to the T effector (TEFF), T circulating memory (TCIRCM), and TRM pools by lineage-tracing and single-cell transcriptome analysis. We demonstrate that a subset of T cell clones possesses a heightened capacity to form TRM, and that enriched expression of TRM-fate-associated genes is already apparent in the circulating TEFF offspring of such clones. In addition, we demonstrate that the capacity to generate TRM is permanently imprinted at the clonal level, before skin entry. Collectively, these data provide compelling evidence for early stage TRM fate decisions and the existence of committed TRM precursor cells in the circulatory TEFF compartment.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Precursor Cells, T-Lymphoid/immunology , Animals , Cell Lineage , Gene Expression Profiling , Mice , Mice, Inbred C57BL
14.
Nat Immunol ; 21(6): 696, 2020 06.
Article in English | MEDLINE | ID: mdl-32210390

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

16.
Pigment Cell Melanoma Res ; 33(2): 334-344, 2020 03.
Article in English | MEDLINE | ID: mdl-31549767

ABSTRACT

NRAS mutations are the most common alterations among RAS isoforms in cutaneous melanoma, with patients harboring these aggressive tumors having a poor prognosis and low survival rate. The main line of treatment for these patients is MAPK pathway-targeted therapies, such as MEK inhibitors, but, unfortunately, the response to these inhibitors is variable due to tumor resistance. Identifying genetic modifiers involved in resistance toward MEK-targeted therapy may assist in the development of new therapeutic strategies, enhancing treatment response and patient survival. Our whole-genome CRISPR-Cas9 knockout screen identified the target Kelch domain-containing F-Box protein 42 (FBXO42) as a factor involved in NRAS-mutant melanoma-acquired resistance to the MEK1/2 inhibitor trametinib. We further show that FBXO42, an E3 ubiquitin ligase, is involved in the TAK1 signaling pathway, possibly prompting an increase in active P38. In addition, we demonstrate that combining trametinib with the TAK1 inhibitor, takinib, is a far more efficient treatment than trametinib alone in NRAS-mutant melanoma cells. Our findings thus show a new pathway involved in NRAS-mutant melanoma resistance and provide new opportunities for novel therapeutic options.


Subject(s)
Drug Resistance, Neoplasm/genetics , F-Box Proteins/genetics , F-Box Proteins/metabolism , GTP Phosphohydrolases/genetics , Genome, Human , Melanoma/genetics , Membrane Proteins/genetics , Skin Neoplasms/genetics , Base Sequence , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Genetic Testing , Humans , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Melanoma/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Models, Biological , Mutation/genetics , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Skin Neoplasms/drug therapy
17.
Cell ; 178(3): 585-599.e15, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31303383

ABSTRACT

New opportunities are needed to increase immune checkpoint blockade (ICB) benefit. Whereas the interferon (IFN)γ pathway harbors both ICB resistance factors and therapeutic opportunities, this has not been systematically investigated for IFNγ-independent signaling routes. A genome-wide CRISPR/Cas9 screen to sensitize IFNγ receptor-deficient tumor cells to CD8 T cell elimination uncovered several hits mapping to the tumor necrosis factor (TNF) pathway. Clinically, we show that TNF antitumor activity is only limited in tumors at baseline and in ICB non-responders, correlating with its low abundance. Taking advantage of the genetic screen, we demonstrate that ablation of the top hit, TRAF2, lowers the TNF cytotoxicity threshold in tumors by redirecting TNF signaling to favor RIPK1-dependent apoptosis. TRAF2 loss greatly enhanced the therapeutic potential of pharmacologic inhibition of its interaction partner cIAP, another screen hit, thereby cooperating with ICB. Our results suggest that selective reduction of the TNF cytotoxicity threshold increases the susceptibility of tumors to immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/drug effects , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Humans , Inhibitor of Apoptosis Proteins/metabolism , Interferon-gamma/metabolism , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred C57BL , Neoplasms/mortality , Neoplasms/therapy , RNA, Guide, Kinetoplastida/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Signal Transduction/drug effects , TNF Receptor-Associated Factor 2/deficiency , TNF Receptor-Associated Factor 2/genetics , Tumor Necrosis Factor-alpha/pharmacology , Interferon gamma Receptor
18.
Nat Immunol ; 20(6): 756-764, 2019 06.
Article in English | MEDLINE | ID: mdl-31110315

ABSTRACT

Emerging data show that tissue-resident memory T (TRM) cells play an important protective role at murine and human barrier sites. TRM cells in the epidermis of mouse skin patrol their surroundings and rapidly respond when antigens are encountered. However, whether a similar migratory behavior is performed by human TRM cells is unclear, as technology to longitudinally follow them in situ has been lacking. To address this issue, we developed an ex vivo culture system to label and track T cells in fresh skin samples. We validated this system by comparing in vivo and ex vivo properties of murine TRM cells. Using nanobody labeling, we subsequently demonstrated in human ex vivo skin that CD8+ TRM cells migrated through the papillary dermis and the epidermis, below sessile Langerhans cells. Collectively, this work allows the dynamic study of resident immune cells in human skin and provides evidence of tissue patrol by human CD8+ TRM cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunologic Memory , Skin/immunology , Animals , Antigens/immunology , Cell Line, Tumor , Cell Movement/immunology , Epidermis/immunology , Epidermis/metabolism , Fluorescent Antibody Technique , Humans , Mice , Organ Specificity/immunology , Single-Domain Antibodies/immunology , Skin/metabolism , Vaccines, DNA/genetics , Vaccines, DNA/immunology
19.
Front Immunol ; 9: 1913, 2018.
Article in English | MEDLINE | ID: mdl-30258433

ABSTRACT

Regulatory B cells (Breg) have been described as a specific immunological subsets in several mouse models. Identification of a human counterpart has remained troublesome, because unique plasma membrane markers or a defining transcription factor have not been identified. Consequently, human Bregs are still primarily defined by production of IL-10. In this study, we sought to elucidate if in vitro-induced human IL-10 producing B cells are a dedicated immunological subset. Using deep immune profiling by multicolor flow cytometry and t-SNE analysis, we show that the majority of cells induced to produce IL-10 co-express pro-inflammatory cytokines IL-6 and/or TNFα. No combination of markers can be identified to define human IL-10+TNFα-IL-6- B cells and rather point to a general activated B cell phenotype. Strikingly, upon culture and restimulation, a large proportion of formerly IL-10 producing B cells lose IL-10 expression, showing that induced IL-10 production is not a stable trait. The combined features of an activated B cell phenotype, transient IL-10 expression and lack of subset-defining markers suggests that in vitro-induced IL-10 producing B cells are not a dedicated subset of regulatory B cells.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Gene Expression Regulation/immunology , Interleukin-10/immunology , Interleukin-6/immunology , Lymphocyte Activation , Tumor Necrosis Factor-alpha/immunology , 3T3 Cells , Animals , B-Lymphocytes, Regulatory/cytology , Female , Flow Cytometry , Humans , Male , Mice
20.
Nat Med ; 24(2): 203-212, 2018 02.
Article in English | MEDLINE | ID: mdl-29334371

ABSTRACT

Intratumor heterogeneity is a key factor contributing to therapeutic failure and, hence, cancer lethality. Heterogeneous tumors show partial therapy responses, allowing for the emergence of drug-resistant clones that often express high levels of the receptor tyrosine kinase AXL. In melanoma, AXL-high cells are resistant to MAPK pathway inhibitors, whereas AXL-low cells are sensitive to these inhibitors, rationalizing a differential therapeutic approach. We developed an antibody-drug conjugate, AXL-107-MMAE, comprising a human AXL antibody linked to the microtubule-disrupting agent monomethyl auristatin E. We found that AXL-107-MMAE, as a single agent, displayed potent in vivo anti-tumor activity in patient-derived xenografts, including melanoma, lung, pancreas and cervical cancer. By eliminating distinct populations in heterogeneous melanoma cell pools, AXL-107-MMAE and MAPK pathway inhibitors cooperatively inhibited tumor growth. Furthermore, by inducing AXL transcription, BRAF/MEK inhibitors potentiated the efficacy of AXL-107-MMAE. These findings provide proof of concept for the premise that rationalized combinatorial targeting of distinct populations in heterogeneous tumors may improve therapeutic effect, and merit clinical validation of AXL-107-MMAE in both treatment-naive and drug-resistant cancers in mono- or combination therapy.


Subject(s)
Immunoconjugates/pharmacology , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Genetic Heterogeneity/drug effects , Humans , Immunoconjugates/immunology , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Mice , Oligopeptides/chemistry , Oligopeptides/immunology , Oligopeptides/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/immunology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/immunology , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/pharmacology , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...