Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
JID Innov ; 3(6): 100221, 2023 Nov.
Article En | MEDLINE | ID: mdl-37731472

Nonkeratinocyte cutaneous malignancies, including breast cancer cutaneous metastasis and melanoma in situ, are often poor surgical candidates. Imiquimod (IMQ), a toll-like receptor 7 agonist that activates innate immunity in the skin, is used to treat these cutaneous malignancies. However, IMQ's modest effect on the activation of adaptive immunity limits its efficacy as a monotherapy. In this study, we demonstrate that topical TSLP cytokine inducers-calcipotriol and retinoic acid-synergize with IMQ to activate CD4+ T-cell immunity against nonkeratinocyte cutaneous malignancies. Topical calcipotriol plus IMQ treatment reduced breast tumor growth compared with calcipotriol or IMQ alone (P < 0.0001). Calcipotriol plus IMQ-mediated tumor suppression was associated with significant infiltration of CD4+ effector T cells in the tumor microenvironment. Notably, topical calcipotriol plus IMQ immunotherapy enabled immune checkpoint blockade therapy to effectively control immunologically cold breast tumors, which was associated with induction of CD4+ T-cell immunity. Topical treatment with calcipotriol plus IMQ and retinoic acid plus IMQ also blocked subcutaneous melanoma growth. These findings highlight the synergistic effect of topical TSLP induction in combination with innate immune cell activation as an effective immunotherapy for malignancies affecting the skin.

2.
Nat Chem Biol ; 17(6): 711-717, 2021 06.
Article En | MEDLINE | ID: mdl-34035522

The zinc-finger transcription factor Helios is critical for maintaining the identity, anergic phenotype and suppressive activity of regulatory T (Treg) cells. While it is an attractive target to enhance the efficacy of currently approved immunotherapies, no existing approaches can directly modulate Helios activity or abundance. Here, we report the structure-guided development of small molecules that recruit the E3 ubiquitin ligase substrate receptor cereblon to Helios, thereby promoting its degradation. Pharmacological Helios degradation destabilized the anergic phenotype and reduced the suppressive activity of Treg cells, establishing a route towards Helios-targeting therapeutics. More generally, this study provides a framework for the development of small-molecule degraders for previously unligandable targets by reprogramming E3 ligase substrate specificity.


DNA-Binding Proteins/drug effects , Ikaros Transcription Factor/drug effects , T-Lymphocytes, Regulatory/drug effects , Transcription Factors/drug effects , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , DNA-Binding Proteins/genetics , Humans , Ikaros Transcription Factor/genetics , Jurkat Cells , Mice , Models, Molecular , Molecular Structure , Mutation/genetics , Small Molecule Libraries , Substrate Specificity , Transcription Factors/genetics , Ubiquitin-Protein Ligases/metabolism
3.
J Invest Dermatol ; 140(7): 1327-1334, 2020 07.
Article En | MEDLINE | ID: mdl-31881212

The skin provides the first line of physical and immunological defense against environmental insults. However, the age-related changes in the immune function of human skin are unclear. Here, we investigated the age-related changes in epidermal Langerhans cells (LCs), which play a sentinel role in the initiation of the immune responses in the skin. We found a significant reduction in the number of epidermal LCs in sun-protected skin with age. Among the possible explanations for this reduction, the number of CD14+ CD207+ CCR6+ dermal-resident monocytes that can differentiate into epidermal LCs was markedly reduced with age (P = 0.0057). Among the chemokines that can recruit these cells into the skin, the expression of CXCL14 was significantly down-regulated in epidermal keratinocytes with age. In addition, we discovered that young skin recruited a significantly higher number of monocytic THP-1 cells compared with old skin ex vivo. This recruitment was blocked by CXCL14 neutralizing antibody and conversely promoted by CXCL14 treatment. Collectively, our findings indicate that decreased CXCL14-mediated recruitment of CD14+ monocytes in human skin results in the reduction of epidermal LCs with age, and CXCL14 may provide a therapeutic target for the prevention of age-related reduction in LCs.


Chemokines, CXC/metabolism , Epidermis/metabolism , Langerhans Cells/cytology , Lipopolysaccharide Receptors/metabolism , Monocytes/cytology , Skin/metabolism , Adult , Age Factors , Aged , Antigens, CD/metabolism , Apoptosis , Cell Count , Cell Movement , Cytokines/metabolism , Dendritic Cells/immunology , Female , Humans , Keratinocytes/drug effects , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Microscopy, Fluorescence , Middle Aged , Receptors, CCR6/metabolism , THP-1 Cells , Young Adult
4.
Nature ; 575(7783): 519-522, 2019 11.
Article En | MEDLINE | ID: mdl-31666702

Immunosuppression increases the risk of cancers that are associated with viral infection1. In particular, the risk of squamous cell carcinoma of the skin-which has been associated with beta human papillomavirus (ß-HPV) infection-is increased by more than 100-fold in immunosuppressed patients2-4. Previous studies have not established a causative role for HPVs in driving the development of skin cancer. Here we show that T cell immunity against commensal papillomaviruses suppresses skin cancer in immunocompetent hosts, and the loss of this immunity-rather than the oncogenic effect of HPVs-causes the markedly increased risk of skin cancer in immunosuppressed patients. To investigate the effects of papillomavirus on carcinogen-driven skin cancer, we colonized several strains of immunocompetent mice with mouse papillomavirus type 1 (MmuPV1)5. Mice with natural immunity against MmuPV1 after colonization and acquired immunity through the transfer of T cells from immune mice or by MmuPV1 vaccination were protected against skin carcinogenesis induced by chemicals or by ultraviolet radiation in a manner dependent on CD8+ T cells. RNA and DNA in situ hybridization probes for 25 commensal ß-HPVs revealed a significant reduction in viral activity and load in human skin cancer compared with the adjacent healthy skin, suggesting a strong immune selection against virus-positive malignant cells. Consistently, E7 peptides from ß-HPVs activated CD8+ T cells from unaffected human skin. Our findings reveal a beneficial role for commensal viruses and establish a foundation for immune-based approaches that could block the development of skin cancer by boosting immunity against the commensal HPVs present in all of our skin.


Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/prevention & control , Papillomaviridae/immunology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Skin Neoplasms/prevention & control , Skin Neoplasms/virology , Symbiosis , Aged , Aged, 80 and over , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinogenesis/radiation effects , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Female , Humans , Immunocompromised Host/immunology , Male , Mice , Middle Aged , Oncogenes , Papillomaviridae/genetics , Papillomaviridae/pathogenicity , RNA, Viral/analysis , RNA, Viral/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Ultraviolet Rays
5.
JCI Insight ; 4(6)2019 03 21.
Article En | MEDLINE | ID: mdl-30895944

BACKGROUND: Topical calcipotriol plus 5-fluorouracil (5-FU) combination is an effective immunotherapy against actinic keratosis (AK), which is a precursor to squamous cell carcinoma (SCC). However, the long-term effectiveness of calcipotriol plus 5-FU treatment for SCC prevention is unknown. METHODS: We performed a blinded prospective cohort study on participants of a randomized double-blind clinical trial in which a 4-day course of topical calcipotriol plus 5-FU combination was compared to Vaseline plus 5-FU (control) for AK treatment. SCC and basal cell carcinoma (BCC) incidences were assessed at 1, 2, and 3 years after trial. Tissues were analyzed for calcipotriol plus 5-FU-induced T cell immunity in the skin. RESULTS: Calcipotriol plus 5-FU-induced tissue-resident memory T (Trm) cell formation in face and scalp skin associated with significantly higher erythema scores compared with control (P < 0.01). Importantly, more participants in the test cohort remained SCC-free over the more than 1,500-day follow-up period (P = 0.0765), and significantly fewer developed SCC on the treated face and scalp within 3 years (2 of 30 [7%] versus 11 of 40 [28%] in control group, hazard ratio 0.215 [95% CI: 0.048-0.972], P = 0.032). Accordingly, significantly more epidermal Trm cells persisted in the calcipotriol plus 5-FU-treated face and scalp skin compared with control (P = 0.0028). There was no significant difference in BCC incidence between the treatment groups. CONCLUSION: A short course of calcipotriol plus 5-FU treatment on the face and scalp is associated with induction of robust T cell immunity and Trm formation against AKs and significantly lowers the risk of SCC development within 3 years of treatment. FUNDING: This research was supported by internal academic funds and by grants from the Burroughs Wellcome Fund, Sidney Kimmel Foundation, Cancer Research Institute, and NIH.


Carcinoma, Squamous Cell/therapy , Immunotherapy/methods , Skin Neoplasms/therapy , Adaptive Immunity , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Calcitriol/therapeutic use , Carcinoma, Basal Cell , Carcinoma, Squamous Cell/pathology , Double-Blind Method , Female , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Humans , Male , Middle Aged , Prospective Studies , Skin/immunology , Skin/pathology , Skin Neoplasms/immunology , Skin Neoplasms/pathology
6.
JAMA Dermatol ; 155(5): 599-603, 2019 May 01.
Article En | MEDLINE | ID: mdl-30785593

IMPORTANCE: Extramammary Paget disease (EMPD), a rare intraepithelial adenocarcinoma, poses a therapeutic challenge with high postoperative recurrence rates and a limited number of effective local treatment options. OBJECTIVE: To describe the use and efficacy of a topical combination of fluorouracil and calcipotriene as a palliative therapy for refractory EMPD. DESIGN, SETTING, AND PARTICIPANTS: This retrospective case series of 3 women with recurrent, refractory EMPD was conducted at Beth Israel Deaconess Medical Center, Boston, Massachusetts and Washington University School of Medicine, St Louis, Missouri. All patients were treated with a 1:1 mixture of fluorouracil, 5%, cream and calcipotriene, 0.005%, cream or ointment. MAIN OUTCOMES AND MEASURES: Clinical and histopathological findings. RESULTS: All 3 women (1 in her 50s, 2 in their 70s) presented with recurrent EMPD (vulvar, perianal, and perioral) after surgery and/or irradiation, and their EMPD was refractory to treatment with imiquimod, 5%, cream. Owing to disease progression and/or intolerable adverse effects from imiquimod, the patients began treatment with a 1:1 mixture of fluorouracil, 5%, cream and calcipotriene, 0.005%, cream. This treatment, which was well tolerated, was followed by clinical improvement in symptoms and appearance of the lesions in all 3 cases and histopathological signs of decreased tumor burden in 2 cases. Patients applied the combination topical therapy to affected areas with differing frequencies, ranging from 1 to 2 days per month to 4 consecutive days every 2 weeks. CONCLUSIONS AND RELEVANCE: Extramammary Paget disease frequently recurs even after aggressive surgical management and can be refractory to many topical and locoregional therapies. Palliative treatment with a combination of fluorouracil and calcipotriene may be a viable option for patients with recurrent, refractory EMPD.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Paget Disease, Extramammary/drug therapy , Palliative Care/methods , Skin Neoplasms/drug therapy , Administration, Topical , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Calcitriol/administration & dosage , Calcitriol/analogs & derivatives , Female , Fluorouracil/administration & dosage , Humans , Imiquimod/administration & dosage , Middle Aged , Paget Disease, Extramammary/pathology , Retrospective Studies , Treatment Outcome
7.
Proc Natl Acad Sci U S A ; 116(7): 2646-2651, 2019 02 12.
Article En | MEDLINE | ID: mdl-30696763

Chronic inflammation's tumor-promoting potential is well-recognized; however, the mechanism underlying the development of this immune environment is unknown. Studying the transition from acute, tumor-suppressive to chronic, tumor-promoting allergic contact dermatitis (ACD) revealed how tumor-promoting chronic inflammation develops. Epidermis-derived interleukin (IL)-33 up-regulation and its induction of regulatory T cell (Treg) accumulation in the skin preceded the transition from acute to chronic ACD and triggered the tumor-promoting immune environment in chronic ACD. Mice lacking IL-33 were protected from chronic ACD and its skin cancer sequela compared with wild-type controls (P = 0.0002). IL-33's direct signaling onto Tregs was required for the development of the tumor-promoting immune environment in the skin. IL-33-Treg signaling was also required for chronic colitis and its associated colorectal cancer development in a colitis model (P < 0.0001). Significantly increased IL-33 and Tregs marked the perilesional skin and colon in patients with cancer-prone chronic inflammatory diseases. Our findings elucidate the role of the IL-33/Treg axis in creating a tumor-promoting immune environment in chronic inflammatory diseases and suggest therapeutic targets for cancer prevention and treatment in high-risk patients.


Colitis/immunology , Colorectal Neoplasms/immunology , Dermatitis, Allergic Contact/immunology , Interleukin-33/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Chronic Disease , Colitis/complications , Colorectal Neoplasms/complications , Dermatitis, Allergic Contact/complications , Humans , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Mice , Mice, Knockout , Skin Neoplasms/complications , Skin Neoplasms/immunology , Up-Regulation
8.
J Invest Dermatol ; 139(1): 62-70, 2019 01.
Article En | MEDLINE | ID: mdl-30030152

High-risk skin cancer is a rare, but severe, complication associated with discoid lupus erythematosus (DLE). Chronic scar, inflammation, UVR, and immunosuppressive medications are proposed explanations for this heightened skin cancer risk; however, the exact mechanism driving skin carcinogenesis in DLE is unknown. The distinct co-localization of multiple independent skin cancers with areas of active inflammation in two DLE patients followed over 8 years strongly suggested that lupus inflammation promotes skin carcinogenesis in DLE. To investigate this clinical observation, we subjected lupus-prone MRL/lpr and control (MRL/n) mice to a skin carcinogenesis protocol. Skin tumors developed preferentially within the cutaneous lupus inflammation without scarring in MRL/lpr mice (P < 0.01). The inflammation in MRL/lpr skin was characterized by the accumulation of regulatory T cells, mast cells, M2 macrophages, and markedly elevated transforming growth factor-ß1 and IL-6 levels, which have been linked to tumor promotion. Tacrolimus treatment reduced skin inflammation and blocked cancer development in MRL/lpr mice (P = 0.0195). A similar tumor-promoting immune environment was detected in SCCs and the perilesional skin of cancer-prone DLE patients. Therefore, discoid lupus inflammation promotes skin cancer in high-risk DLE patients, and blocking the inflammation may be critical for preventing this life-threatening complication of DLE.


Cytokines/metabolism , Inflammation/pathology , Lupus Erythematosus, Discoid/pathology , Skin Neoplasms/etiology , Skin/pathology , Animals , Carcinogenesis , Chronic Disease , Female , Humans , Inflammation/complications , Inflammation/metabolism , Lupus Erythematosus, Discoid/complications , Lupus Erythematosus, Discoid/metabolism , Mice , Mice, Inbred MRL lpr , Middle Aged , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
9.
J Clin Invest ; 126(4): 1458-70, 2016 Apr 01.
Article En | MEDLINE | ID: mdl-26927668

Advances in the field of cancer immunology, including studies on tumor-infiltrating CD8+ cytotoxic T lymphocytes (CTLs), have led to new immunotherapeutics with proven efficacy against late-stage cancers. However, the antitumor potential of the immune system in targeting early-stage cancers remains uncertain. Here, we demonstrated that both genetic and chemical induction of thymic stromal lymphopoietin (TSLP) at a distant site leads to robust antitumor immunity against spontaneous breast carcinogenesis in mice. Breast tumors exposed to high circulating levels of TSLP were arrested at an early adenoma-like stage and were prevented from advancing to late carcinoma and metastasis. Additionally, CD4+ Th2 cells mediated the antitumor effects of TSLP, challenging the notion that Th2 cells only promote cancer. We also discovered that TSLP is expressed by the breast tumor cells themselves and acts to block breast cancer promotion. Moreover, TSLP-induced immunity also blocked early stages of pancreatic cancer development. Together, our findings demonstrate that TSLP potently induces immunity directed against early stages of breast cancer development without causing inflammation in the normal breast tissue. Moreover, our results highlight a previously unappreciated function of the immune system in controlling the early development of cancer and establish a fundamental role for TSLP and Th2 cells in tumor immunity against early-stage cancers.


CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Immunity, Cellular , Mammary Neoplasms, Experimental/immunology , Th2 Cells/immunology , Tumor Suppressor Proteins/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Cytokines/genetics , Female , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Th2 Cells/pathology , Tumor Suppressor Proteins/genetics , Thymic Stromal Lymphopoietin
...