Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
1.
J Orthop Sci ; 2023 Oct 14.
Article in English | MEDLINE | ID: mdl-37845161

ABSTRACT

BACKGROUND: Dropped head syndrome (DHS) is followed by severe cervical extension muscle weakness that results in chin-on chest deformity. However, maintaining a neutral cervical position can be temporarily possible, and the diagnosis of DHS might sometimes be difficult. The purpose of the present study is to examine a novel clinical test (DHS test) as the diagnostic utility for objective evaluation that focuses on cervical extension condition in the prone position. METHODS: One hundred subjects were diagnosed with isolated neck extensor myopathy (INEM)-DHS at our hospital (17 men and 83 women, mean age 75.0 ± 8.5 years), and 62 subjects were enrolled as age-matched controls. The DHS test consisted of three examinations; the first was "Ceiling gazing test" in standing position, the second was horizontal gazing in "Sphinx prone position test", and the third was horizontal gazing in "Hands and knees prone position test". We investigated the sensitivity and specificity of the DHS test for DHS. RESULTS: The patients showing positive in the INEM-DHS group were 63/100 in Ceiling gaze test, 73/100 in the Sphinx prone position test, and 91/100 in the Hands and knees prone position test. In the control group, 0/62 patients presented positive in the Ceiling gaze test, 4/62 in the Sphinx prone position test, and 0/62 in the Hands and knees prone position test. Sensitivity and specificity of the DHS test were 63.0%/100%, 73.0%/93.5%, and 91.0%/100% in the Ceiling gaze test, Sphinx position prone position test, and Hands and knees prone position test, respectively. CONCLUSION: The prone position cervical extension test (DHS test) would be useful as a novel objective diagnostic tool for INEM-DHS.

2.
Int J Hyperthermia ; 40(1): 2263672, 2023.
Article in English | MEDLINE | ID: mdl-37806666

ABSTRACT

Mechanical high-intensity focused ultrasound (M-HIFU), which includes histotripsy, is a non-ionizing, non-thermal ablation technology that can be delivered by noninvasive methods. Because acoustic cavitation is the primary mechanism of tissue disruption, histotripsy is distinct from the conventional HIFU techniques resulting in hyperthermia and thermal injury. Phase I human trials have shown the initial safety and efficacy of histotripsy in treating patients with malignant liver tumors. In addition to tissue ablation, a promising benefit of M-HIFU has been stimulating a local and systemic antitumor immune response in preclinical models and potentially in the Phase I trial. Preclinical studies combining systemic immune therapies appear promising, but clinical studies of combinations have been complicated by systemic toxicities. Consequently, combining M-HIFU with systemic immunotherapy has been demonstrated in preclinical models and may be testing in future clinical studies. An additional alternative is to combine intratumoral M-HIFU and immunotherapy using microcatheter-placed devices to deliver both M-HIFU and immunotherapy intratumorally. The promise of M-HIFU as a component of anti-cancer therapy is promising, but as forms of HIFU are tested in preclinical and clinical studies, investigators should report not only the parameters of the energy delivered but also details of the preclinical models to enable analysis of the immune responses. Ultimately, as clinical trials continue, clinical responses and immune analysis of patients undergoing M-HIFU including forms of histotripsy will provide opportunities to optimize clinical responses and to optimize application and scheduling of M-HIFU in the context of the multi-modality care of the cancer patient.


Subject(s)
Carcinoma, Hepatocellular , High-Intensity Focused Ultrasound Ablation , Liver Neoplasms , Humans , High-Intensity Focused Ultrasound Ablation/methods , Immunotherapy
3.
Cancer Gene Ther ; 30(6): 803-811, 2023 06.
Article in English | MEDLINE | ID: mdl-36765179

ABSTRACT

Therapeutic cancer vaccines, designed to activate immune effectors against tumor antigens, utilize a number of different platforms for antigen delivery. Among these are messenger RNAs (mRNA), successfully deployed in some prophylactic SARS-CoV2 vaccines. To enhance the immunogenicity of mRNA-delivered epitopes, self-replicating RNAs (srRNA) that markedly increase epitope expression have been developed. These vectors are derived from positive-strand RNA viruses in which the structural protein genes have been replaced with heterologous genes of interest, and the structural proteins are provided in trans to create single cycle viral replicon particles (VRPs). Clinical stage srRNA vectors have been derived from alphaviruses, including Venezuelan Equine Encephalitis (VEE), Sindbis, and Semliki Forest virus (SFV) and have encoded the tumor antigens carcinoembryonic antigen (CEA), human epidermal growth factor receptor 2 (HER2), prostate specific membrane antigen (PSMA), and human papilloma virus (HPV) antigens E6 and E7. Adverse events have mainly been grade 1 toxicities and minimal injection site reactions. We review here the clinical experience with these vaccines and our recent safety data from a study combining a VRP encoding HER2 plus an anti-PD1 monoclonal antibody (pembrolizumab). This experience with VRP-based srRNA supports recent development of fully synthetic srRNA technologies, where the viral structural proteins are replaced with protective lipid nanoparticles (LNP), cationic nanoemulsions or polymers.


Subject(s)
COVID-19 , Cancer Vaccines , Encephalitis Virus, Venezuelan Equine , Neoplasms , Humans , RNA, Viral/genetics , Cancer Vaccines/genetics , Encephalitis Virus, Venezuelan Equine/genetics , COVID-19/genetics , SARS-CoV-2/genetics , RNA, Messenger , Replicon , Genetic Vectors , Neoplasms/genetics , Neoplasms/therapy
4.
Cancers (Basel) ; 14(23)2022 Nov 23.
Article in English | MEDLINE | ID: mdl-36497243

ABSTRACT

Ductal carcinoma in situ (DCIS) of the breast is often managed by lumpectomy and radiation or mastectomy, despite its indolent features. Effective non-invasive treatment strategies could reduce the morbidity of DCIS treatment. We have exploited the high heat shock protein 90 (HSP90) activity in premalignant and malignant breast disease to non-invasively detect and selectively ablate tumors using photodynamic therapy (PDT). PDT with the HSP90-targeting photosensitizer, HS201, can not only ablate invasive breast cancers (BCs) while sparing non-tumor tissue, but also induce antitumor immunity. We hypothesized that HS201-PDT would both non-invasively ablate DCIS and prevent progression to invasive BC. We tested in vitro selective uptake and photosensitivity of HS201 in DCIS cell lines compared to the non-selective parental verteporfin, and assessed in vivo antitumor efficacy in mammary fat pad and intraductal implantation models. Selective uptake of HS201 enabled treatment of intraductal lesions while minimizing toxicity to non-tumor tissue. The in vivo activity of HS201-PDT was also tested in female MMTV-neu mice prior to the development of spontaneous invasive BC. Mice aged 5 months were administered HS201, and their mammary glands were exposed to laser light. HS201-PDT delayed the emergence of invasive BC, significantly prolonged disease-free survival (DFS) (p = 0.0328) and tended to improve overall survival compared to the no-treatment control (p = 0.0872). Systemic administration of anti-PD-L1 was combined with HS201-PDT and was tested in a more aggressive spontaneous tumor model, HER2delta16 transgenic mice. A single PDT dose combined with anti-PD-L1 improved DFS compared to the no-treatment control, which was significantly improved with repetitive HS201-PDT given with anti-PD-L1 (p = 0.0319). In conclusion, a non-invasive, skin- and tissue-sparing PDT strategy in combination with anti-PD-L1 antibodies effectively prevented malignant progression of DCIS to invasive BC. This non-invasive treatment strategy of DCIS may be safe and effective, while providing an option to reduce the morbidity of current conventional treatment for patients with DCIS. Clinical testing of HS201 is currently underway.

5.
J Immunother Cancer ; 10(9)2022 09.
Article in English | MEDLINE | ID: mdl-36171008

ABSTRACT

BACKGROUND: We previously demonstrated potent antitumor activity against human breast cancer xenografts using photodynamic therapy (PDT) targeting a novel tumor-specific photosensitizer (HS201), which binds heat shock protein 90 (HS201-PDT). However, induction of systemic antitumor immunity by HS201-PDT alone or by the combination strategy with immune checkpoint blockade has yet to be determined. METHODS: Using unilateral and bilateral implantation models of syngeneic breast tumors (E0771, MM3MG-HER2, and JC-HER3) in mice, we assessed whether HS201-PDT could induce local and systemic antitumor immunity. In an attempt to achieve a stronger abscopal effect for distant tumors, the combination strategy with anti-PD-L1 antibody was tested. Tumor-infiltrating leukocytes were analyzed by single cell RNA-sequencing and receptor-ligand interactome analysis to characterize in more detailed the mechanisms of action of the treatment and key signaling pathways involved. RESULTS: HS201-PDT demonstrated greater tumor control and survival in immune competent mice than in immunocompromised mice, suggesting the role of induced antitumor immunity; however, survival was modest and an abscopal effect on distant implanted tumor was weak. A combination of HS201-PDT with anti-PD-L1 antibody demonstrated the greatest antigen-specific immune response, tumor growth suppression, prolonged mouse survival time and abscopal effect. The most significant increase of intratumoral, activated CD8+T cells and decrease of exhausted CD8+T cells occurred following combination treatment compared with HS201-PDT monotherapy. Receptor-ligand interactome analysis showed marked enhancement of several pathways, such as CXCL, GALECTIN, GITRL, PECAM1 and NOTCH, associated with CD8+T cell activation in the combination group. Notably, the expression of the CXCR3 gene signature was the highest in the combination group, possibly explaining the enhanced tumor infiltration by T cells. CONCLUSIONS: The increased antitumor activity and upregulated CXCR3 gene signature induced by the combination of anti-PD-L1 antibody with HS201-PDT warrants the clinical testing of HS201-PDT combined with PD-1/PD-L1 blockade in patients with breast cancer, and the use of the CXCR3 gene signature as a biomarker.


Subject(s)
Breast Neoplasms , Photochemotherapy , Animals , Cell Line, Tumor , Female , Galectins , Heat-Shock Proteins , Humans , Immune Checkpoint Inhibitors , Ligands , Mice , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Programmed Cell Death 1 Receptor , RNA
6.
J Immunother Cancer ; 10(3)2022 03.
Article in English | MEDLINE | ID: mdl-35361727

ABSTRACT

BACKGROUND: The majority of colorectal carcinomas (CRCs) are insensitive to programmed death protein-1/programmed death-ligand 1 (anti-PD-1/PD-L1) immune checkpoint inhibitor (ICI) antibodies. While there are many causes for ICI insensitivity, recent studies suggest that suppression of innate immune gene expression in tumor cells could be a root cause of this insensitivity and an important factor in the evolution of tumor immunosuppression. METHODS: We first assessed the reduction of mitochondrial antiviral signaling gene (MAVS) and related RIG-I pathway gene expression in several patient RNA expression datasets. We then engineered MAVS expressing tumor cells and tested their ability to elicit innate and adaptive anti-tumor immunity using both in vitro and in vivo approaches, which we then confirmed using MAVS expressing viral vectors. Finally, we observed that MAVS stimulated PD-L1 expression in multiple cell types and then assessed the combination of PD-L1 ICI antibodies with MAVS tumor expression in vivo. RESULTS: MAVS was significantly downregulated in CRCs, but its re-expression could stimulate broad cellular interferon-related responses, in both murine and patient-derived CRCs. In vivo, local MAVS expression elicited significant anti-tumor responses in both immune-sensitive and insensitive CRC models, through the stimulation of an interferon responsive axis that provoked tumor antigen-specific adaptive immunity. Critically, we found that tumor-intrinsic MAVS expression triggered systemic adaptive immune responses that enabled abscopal CD8 +T cell cytotoxicity against distant CRCs. As MAVS also induced PD-L1 expression, we further found synergistic anti-tumor responses in combination with anti-PD-L1 ICIs. CONCLUSION: These data demonstrate that intratumoral MAVS expression results in local and systemic tumor antigen-specific T cell responses, which could be combined with PD-L1 ICI to permit effective anti-tumor immunotherapy in ICI resistant cancers.


Subject(s)
Colorectal Neoplasms , Immune Checkpoint Inhibitors , Animals , Antiviral Agents , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Mice , Signal Transduction
7.
J Immunother Cancer ; 10(1)2022 01.
Article in English | MEDLINE | ID: mdl-35039461

ABSTRACT

BACKGROUND: Despite multimodal adjuvant management with radiotherapy, chemotherapy and hormonal therapies, most surgically resected primary breast cancers relapse or metastasize. A potential solution to late and distant recurrence is to augment systemic antitumor immunity, in part by appropriately presenting tumor antigens, but also by modulating the immunosuppressive tumor microenvironment (TME). We previously validated this concept in models of murine carcinoma treated with a novel predominately microcavitating version of high-intensity focused ultrasound (HIFU), mechanical high-intensity focused ultrasound (M-HIFU). Here we elucidated the mechanisms of enhanced antitumor immunity by M-HIFU over conventional thermal high-intensity focused ultrasound (T-HIFU) and investigated the potential of the combinatorial strategy with an immune checkpoint inhibitor, anti-PD-L1 antibody. METHODS: The antitumor efficacy of treatments was investigated in syngeneic murine breast cancer models using triple-negative (E0771) or human ErbB-2 (HER2) expressing (MM3MG-HER2) tumors in C57BL/6 or BALB/c mice, respectively. Induction of systemic antitumor immunity by the treatments was tested using bilateral tumor implantation models. Flow cytometry, immunohistochemistry, and single-cell RNA sequencing were performed to elucidate detailed effects of HIFU treatments or combination treatment on TME, including the activation status of CD8 T cells and polarization of tumor-associated macrophages (TAMs). RESULTS: More potent systemic antitumor immunity and tumor growth suppression were induced by M-HIFU compared with T-HIFU. Molecular characterization of the TME after M-HIFU by single-cell RNA sequencing demonstrated repolarization of TAM to the immunostimulatory M1 subtype compared with TME post-T-HIFU. Concurrent anti-PD-L1 antibody administration or depletion of CD4+ T cells containing a population of regulatory T cells markedly increased T cell-mediated antitumor immunity and tumor growth suppression at distant, untreated tumor sites in M-HIFU treated mice compared with M-HIFU monotherapy. CD8 T and natural killer cells played major roles as effector cells in the combination treatment. CONCLUSIONS: Physical disruption of the TME by M-HIFU repolarizes TAM, enhances T-cell infiltration, and, when combined with anti-PD-L1 antibody, mediates superior systemic antitumor immune responses and distant tumor growth suppression. These findings suggest M-HIFU combined with anti-PD-L1 may be useful in reducing late recurrence or metastasis when applied to primary tumors.


Subject(s)
Combined Modality Therapy/methods , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Ultrasonography/methods , Animals , Cell Line, Tumor , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Mice , Tumor Microenvironment
8.
Mol Cancer Ther ; 21(1): 217-226, 2022 01.
Article in English | MEDLINE | ID: mdl-34675120

ABSTRACT

A noninvasive test to discriminate indolent prostate cancers from lethal ones would focus treatment where necessary while reducing overtreatment. We exploited the known activity of heat shock protein 90 (Hsp90) as a chaperone critical for the function of numerous oncogenic drivers, including the androgen receptor and its variants, to detect aggressive prostate cancer. We linked a near-infrared fluorescing molecule to an HSP90 binding drug and demonstrated that this probe (designated HS196) was highly sensitive and specific for detecting implanted prostate cancer cell lines with greater uptake by more aggressive subtypes. In a phase I human study, systemically administered HS196 could be detected in malignant nodules within prostatectomy specimens. Single-cell RNA sequencing identified uptake of HS196 by malignant prostate epithelium from the peripheral zone (AMACR+ERG+EPCAM+ cells), including SYP+ neuroendocrine cells that are associated with therapeutic resistance and metastatic progression. A theranostic version of this molecule is under clinical testing.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/genetics , Animals , Cell Line, Tumor , Humans , Male , Mice , Mice, SCID , Prostatic Neoplasms/pathology
9.
Clin Cancer Res ; 27(9): 2481-2493, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33593880

ABSTRACT

PURPOSE: Triple-negative breast cancer (TNBC) is an aggressive disease with limited therapeutic options. Antibodies targeting programmed cell death protein 1 (PD-1)/PD-1 ligand 1 (PD-L1) have entered the therapeutic landscape in TNBC, but only a minority of patients benefit. A way to reliably enhance immunogenicity, T-cell infiltration, and predict responsiveness is critically needed. PATIENTS AND METHODS: Using mouse models of TNBC, we evaluate immune activation and tumor targeting of intratumoral IL12 plasmid followed by electroporation (tavokinogene telseplasmid; Tavo). We further present a single-arm, prospective clinical trial of Tavo monotherapy in patients with treatment refractory, advanced TNBC (OMS-I140). Finally, we expand these findings using publicly available breast cancer and melanoma datasets. RESULTS: Single-cell RNA sequencing of murine tumors identified a CXCR3 gene signature (CXCR3-GS) following Tavo treatment associated with enhanced antigen presentation, T-cell infiltration and expansion, and PD-1/PD-L1 expression. Assessment of pretreatment and posttreatment tissue from patients confirms enrichment of this CXCR3-GS in tumors from patients that exhibited an enhancement of CD8+ T-cell infiltration following treatment. One patient, previously unresponsive to anti-PD-L1 therapy, but who exhibited an increased CXCR3-GS after Tavo treatment, went on to receive additional anti-PD-1 therapy as their immediate next treatment after OMS-I140, and demonstrated a significant clinical response. CONCLUSIONS: These data show a safe, effective intratumoral therapy that can enhance antigen presentation and recruit CD8 T cells, which are required for the antitumor efficacy. We identify a Tavo treatment-related gene signature associated with improved outcomes and conversion of nonresponsive tumors, potentially even beyond TNBC.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Drug Resistance, Neoplasm/genetics , Interleukin-12/genetics , Plasmids/administration & dosage , Receptors, CXCR3/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/therapy , Animals , Cell Line, Tumor , Disease Management , Disease Models, Animal , Electroporation , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunophenotyping , Injections, Intralesional , Iron Compounds , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Melanoma/therapy , Mice , Plasmids/genetics , Treatment Outcome , Triple Negative Breast Neoplasms/etiology , Triple Negative Breast Neoplasms/pathology
11.
J Immunother Cancer ; 8(2)2020 11.
Article in English | MEDLINE | ID: mdl-33177177

ABSTRACT

BACKGROUND: There remains a significant need to eliminate the risk of recurrence of resected cancers. Cancer vaccines are well tolerated and activate tumor-specific immune effectors and lead to long-term survival in some patients. We hypothesized that vaccination with alphaviral replicon particles encoding tumor associated antigens would generate clinically significant antitumor immunity to enable prolonged overall survival (OS) in patients with both metastatic and resected cancer. METHODS: OS was monitored for patients with stage IV cancer treated in a phase I study of virus-like replicon particle (VRP)-carcinoembryonic antigen (CEA), an alphaviral replicon particle encoding a modified CEA. An expansion cohort of patients (n=12) with resected stage III colorectal cancer who had completed their standard postoperative adjuvant chemotherapy was administered VRP-CEA every 3 weeks for a total of 4 immunizations. OS and relapse-free survival (RFS) were determined, as well as preimmunization and postimmunization cellular and humoral immunity. RESULTS: Among the patients with stage IV cancer, median follow-up was 10.9 years and 5-year survival was 17%, (95% CI 6% to 33%). Among the patients with stage III cancer, the 5-year RFS was 75%, (95%CI 40% to 91%); no deaths were observed. At a median follow-up of 5.8 years (range: 3.9-7.0 years) all patients were still alive. All patients demonstrated CEA-specific humoral immunity. Patients with stage III cancer had an increase in CD8 +TEM (in 10/12) and decrease in FOXP3 +Tregs (in 10/12) following vaccination. Further, CEA-specific, IFNγ-producing CD8+granzyme B+TCM cells were increased. CONCLUSIONS: VRP-CEA induces antigen-specific effector T cells while decreasing Tregs, suggesting favorable immune modulation. Long-term survivors were identified in both cohorts, suggesting the OS may be prolonged.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colonic Neoplasms/drug therapy , Immunologic Memory/physiology , T-Lymphocytes, Regulatory/immunology , Colonic Neoplasms/mortality , Female , Humans , Male , Neoplasm Staging , Survival Analysis
12.
Commun Biol ; 3(1): 226, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32385408

ABSTRACT

Photodynamic therapy (PDT) ablates malignancies by applying focused near-infrared (nIR) light onto a lesion of interest after systemic administration of a photosensitizer (PS); however, the accumulation of existing PS is not tumor-exclusive. We developed a tumor-localizing strategy for PDT, exploiting the high expression of heat shock protein 90 (Hsp90) in cancer cells to retain high concentrations of PS by tethering a small molecule Hsp90 inhibitor to a PS (verteporfin, VP) to create an Hsp90-targeted PS (HS201). HS201 accumulates to a greater extent than VP in breast cancer cells both in vitro and in vivo, resulting in increased treatment efficacy of HS201-PDT in various human breast cancer xenografts regardless of molecular and clinical subtypes. The therapeutic index achieved with Hsp90-targeted PDT would permit treatment not only of localized tumors, but also more diffusely infiltrating processes such as inflammatory breast cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Photochemotherapy/statistics & numerical data , Photosensitizing Agents/administration & dosage , Verteporfin/administration & dosage , Cell Line, Tumor , HSP90 Heat-Shock Proteins/administration & dosage , HSP90 Heat-Shock Proteins/radiation effects , Humans , MCF-7 Cells
13.
Immunotherapy ; 11(11): 953-966, 2019 08.
Article in English | MEDLINE | ID: mdl-31192764

ABSTRACT

Aim: The purpose of this study was to determine whether addition of anti-PD-1 antibody increased the immunogenicity and anti-tumor activity of Ad-CEA vaccination in a murine model of colon cancer. Methods: Ad-CEA was administered prior to implantation of MC-38-CEA cells followed by administration of anti-PD-1 antibody. CEA-specific T-cell responses were measured by flow cytometry and ELISPOT. Dynamic co-culture of splenocytes with tumor cells was conducted to analyze anti-tumor activities. Tumor infiltration by lymphocytes was measured by IHC. Tumor volume and overall survival were also recorded. Results: Ad-CEA combined with anti-PD-1 antibody showed greater anti-tumor activity compared with either alone. The combination also increased T-cell infiltration but decreased Tregs. Conclusion: Combining Ad-CEA vaccination with anti-PD-1 antibody enhanced anti-tumor activity and immune responses.


Subject(s)
Adenoviridae , Antineoplastic Agents, Immunological/pharmacology , Cancer Vaccines , Carcinoembryonic Antigen , Colonic Neoplasms , Neoplasms, Experimental , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Vaccination , Animals , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Female , Humans , Mice , NIH 3T3 Cells , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Programmed Cell Death 1 Receptor/immunology
14.
Eur Arch Otorhinolaryngol ; 276(7): 2055-2060, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31076880

ABSTRACT

PURPOSE: We morphometrically analyzed human facial muscles, and evaluated the Yanagihara facial nerve grading system using our data. METHODS: We used 15 types of human facial muscle, 2 types of masticatory muscle and 2 types of skeletal muscle. The materials were obtained from 11 Japanese male cadavers aged 43-86 years. We counted the muscle fibers and measured the transverse area of the muscle fibers (TAMF), and then calculated the number of muscle fibers (NMF) per mm2 and the average TAMF. RESULTS: We found a significant correlation between average TAMF and NMF (r = - 0.70; p < 0.01). We classified facial muscles into three types based on the correlational results. Type A had a low average TAMF and high NMF. Type C had a high average TAMF and low NMF. Masticatory and skeletal muscles were characterized as Type C. Type B was intermediate between Types A and C. CONCLUSIONS: Pathological changes in the facial muscles in facial nerve palsy seem to vary according to the type of facial muscle, because each facial muscle has a unique fiber-type composition. As the nine discrete facial expressive states evaluated in the Yanagihara system involve all three facial muscle types of our classification, the Yanagihara system is an outstanding system for grading facial nerve palsy in terms of the facial muscle morphology.


Subject(s)
Facial Muscles , Facial Nerve/pathology , Facial Paralysis , Adult , Aged , Cadaver , Face , Facial Muscles/innervation , Facial Muscles/pathology , Facial Paralysis/classification , Facial Paralysis/pathology , Humans , Male , Middle Aged , Pathology, Clinical/methods
15.
Clin Cancer Res ; 25(9): 2725-2736, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30635338

ABSTRACT

PURPOSE: Immune-based therapy for metastatic breast cancer has had limited success, particularly in molecular subtypes with low somatic mutations rates. Strategies to augment T-cell infiltration of tumors include vaccines targeting established oncogenic drivers such as the genomic amplification of HER2. We constructed a vaccine based on a novel alphaviral vector encoding a portion of HER2 (VRP-HER2). PATIENTS AND METHODS: In preclinical studies, mice were immunized with VRP-HER2 before or after implantation of hHER2+ tumor cells and HER2-specific immune responses and antitumor function were evaluated. We tested VRP-HER2 in a phase I clinical trial where subjects with advanced HER2-overexpressing malignancies in cohort 1 received VRP-HER2 every 2 weeks for a total of 3 doses. In cohort 2, subjects received the same schedule concurrently with a HER2-targeted therapy. RESULTS: Vaccination in preclinical models with VRP-HER2 induced HER2-specific T cells and antibodies while inhibiting tumor growth. VRP-HER2 was well tolerated in patients and vaccination induced HER2-specific T cells and antibodies. Although a phase I study, there was 1 partial response and 2 patients with continued stable disease. Median OS was 50.2 months in cohort 1 (n = 4) and 32.7 months in cohort 2 (n = 18). Perforin expression by memory CD8 T cells post-vaccination significantly correlated with improved PFS. CONCLUSIONS: VRP-HER2 increased HER2-specific memory CD8 T cells and had antitumor effects in preclinical and clinical studies. The expansion of HER2-specific memory CD8 T cells in vaccinated patients was significantly correlated with increased PFS. Subsequent studies will seek to enhance T-cell activity by combining with anti-PD-1.


Subject(s)
Breast Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Immunity, Humoral/immunology , Immunologic Memory/immunology , Receptor, ErbB-2/immunology , Vaccines, Subunit/administration & dosage , Adult , Aged , Alphavirus/genetics , Animals , Apoptosis , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cancer Vaccines/immunology , Cell Proliferation , Dendritic Cells/immunology , Female , Follow-Up Studies , Genetic Vectors/administration & dosage , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , Receptor, ErbB-2/metabolism , Survival Rate , Tumor Cells, Cultured , Vaccines, Subunit/immunology , Xenograft Model Antitumor Assays
16.
Biochem J ; 476(3): 535-546, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30635359

ABSTRACT

The Wnt signaling pathway, known for regulating genes critical to normal embryonic development and tissue homeostasis, is dysregulated in many types of cancer. Previously, we identified that the anthelmintic drug niclosamide inhibited Wnt signaling by promoting internalization of Wnt receptor Frizzled 1 and degradation of Wnt signaling pathway proteins, Dishevelled 2 and ß-catenin, contributing to suppression of colorectal cancer growth in vitro and in vivo Here, we provide evidence that niclosamide-mediated inhibition of Wnt signaling is mediated through autophagosomes induced by niclosamide. Specifically, niclosamide promotes the co-localization of Frizzled 1 or ß-catenin with LC3, an autophagosome marker. Niclosamide inhibition of Wnt signaling is attenuated in autophagosome-deficient ATG5-/- MEF cells or cells expressing shRNA targeting Beclin1, a critical constituent of autophagosome. Treatment with the autophagosome inhibitor 3MA blocks niclosamide-mediated Frizzled 1 degradation. The sensitivity of colorectal cancer cells to growth inhibition by niclosamide is correlated with autophagosome formation induced by niclosamide. Niclosamide inhibits mTORC1 and ULK1 activities and induces LC3B expression in niclosamide-sensitive cell lines, but not in the niclosamide-resistant cell lines tested. Interestingly, niclosamide is a less effective inhibitor of Wnt-responsive genes (ß-catenin, c-Myc, and Survivin) in the niclosamide-resistant cells than in the niclosamide-sensitive cells, suggesting that deficient autophagy induction by niclosamide compromises the effect of niclosamide on Wnt signaling. Our findings provide a mechanistic understanding of the role of autophagosomes in the inhibition of Wnt signaling by niclosamide and may provide biomarkers to assist selection of patients whose tumors are likely to respond to niclosamide.


Subject(s)
Autophagy/drug effects , Colorectal Neoplasms/drug therapy , Niclosamide/pharmacology , Wnt Signaling Pathway/drug effects , Autophagy/genetics , Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dishevelled Proteins/genetics , Dishevelled Proteins/metabolism , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , HCT116 Cells , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
17.
Clin Cancer Res ; 25(1): 9-11, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30377197

ABSTRACT

Systemic IL12 therapy has potent antitumor effects, but clinical delivery of this potent cytokine has been complicated by systemic toxicity. A novel strategy to deliver IL12 to the tumor microenvironment appears promising in a first-in-human study, appearing to stimulate tumor-specific adaptive immune responses with minimal systemic toxicity.See related article by Strauss et al., p. 99.


Subject(s)
Neoplasms , State Medicine , Cytokines , Humans , Interleukin-12 , Tumor Microenvironment
18.
Adv Exp Med Biol ; 1072: 91-96, 2018.
Article in English | MEDLINE | ID: mdl-30178329

ABSTRACT

The aim of this study was to test the effects of aerobic cycling training on O2 dynamics in several leg muscles in early post-myocardial infarction (post-MI). Fifteen post-MI patients were divided into a 12-week training group (TR, n = 9) or a control/non-training group (CON, n = 6). All participants performed ramp bicycle exercise until exhaustion at two times: within 12-35 days of their MI and then again 12 weeks later. Muscle O2 saturation (SmO2) and total hemoglobin concentration (∆total-Hb) were monitored continuously at thigh and lower leg muscles by near infrared spectroscopy. In CON, there were no significant alterations in muscle O2 dynamics between before and after 12 weeks at any measurement sites. In TR, after 12 weeks, lower SmO2 was observed at all measurement sites. In total-Hb, no significant changes were found after training at any measurement sites in TR. Moreover, the muscle deoxygenation after 12 weeks was related to an improvement of peak O2 uptake in all muscles. Our findings suggest that aerobic cycling training may be useful for early post-MI patients to improve peak aerobic capacity via enhancement of muscle deoxygenation and O2 extraction at several leg muscles.


Subject(s)
Exercise Therapy/methods , Exercise/physiology , Muscle, Skeletal/metabolism , Myocardial Infarction/rehabilitation , Oxygen Consumption/physiology , Aged , Female , Humans , Male , Middle Aged
19.
Breast Cancer Res ; 20(1): 90, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30092835

ABSTRACT

BACKGROUND: Upregulation of human epidermal growth factor receptor 3 (HER3) is a major mechanism of acquired resistance to therapies targeting its heterodimerization partners epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2), but also exposes HER3 as a target for immune attack. We generated an adenovirus encoding full length human HER3 (Ad-HER3) to serve as a cancer vaccine. Previously we reported the anti-tumor efficacy and function of the T cell response to this vaccine. We now provide a detailed assessment of the antitumor efficacy and functional mechanisms of the HER3 vaccine-induced antibodies (HER3-VIAs) in serum from mice immunized with Ad-HER3. METHODS: Serum containing HER3-VIA was tested in complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC) assays and for its effect on HER3 internalization and degradation, downstream signaling of HER3 heterodimers and growth of metastatic HER2+ (BT474M1), HER2 therapy-resistant (rBT474), and triple negative (MDA-MB-468) breast cancers. RESULTS: HER3-VIAs mediated CDC and ADCC, HER3 internalization, interruption of HER3 heterodimer-driven tumor signaling pathways, and anti-proliferative effects against HER2+ tumor cells in vitro and significant antitumor effects against metastatic HER2+ BT474M1, treatment refractory HER2+ rBT474 and triple negative MDA-MB-468 in vivo. CONCLUSIONS: In addition to the T cell anti-tumor response induced by Ad-HER3, the HER3-VIAs provide additional functions to eliminate tumors in which HER3 signaling mediates aggressive behavior or acquired resistance to HER2-targeted therapy. These data support clinical studies of vaccination against HER3 prior to or concomitantly with other therapies to prevent outgrowth of therapy-resistant HER2+ and triple negative clones.


Subject(s)
Antibodies/immunology , Antineoplastic Agents/pharmacology , Cancer Vaccines/immunology , Receptor, ErbB-3/immunology , Triple Negative Breast Neoplasms/therapy , Adenoviridae/genetics , Animals , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents/therapeutic use , Breast/pathology , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Epitope Mapping , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Immunization, Passive/methods , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
20.
Clin Cancer Res ; 23(24): 7531-7542, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-28993342

ABSTRACT

Purpose: Hsp90, a chaperone to numerous molecular pathways in malignant cells, is elevated in aggressive breast cancers. We hypothesized that identifying breast cells with elevated Hsp90 activity in situ could result in early detection of aggressive breast cancers.Experimental Design: We exploited the uptake of an Hsp90 inhibitor by malignant cells to create an imaging probe (HS131) of Hsp90 activity by linking it to a near-infrared (nIR) dye. HS131 uptake into cells correlated with cell membrane expression of Hsp90 and was used to image molecular subtypes of murine and human breast cancers in vitro and in murine models.Results: HS131 imaging was both sensitive and specific in detecting the murine 4T1 breast cancer cell line, as well as subclones with differing metastatic potential. Highly metastatic subclones (4T07) had high HS131 uptake, but subclones with lower metastatic potential (67NR, 168FARN) had low HS131 uptake. We generated isogenic cell lines to demonstrate that overexpression of a variety of specific oncogenes resulted in high HS131 uptake and retention. Finally, we demonstrated that HS131 could be used to detect spontaneous tumors in MMTV-neu mice, as well as primary and metastatic human breast cancer xenografts. HS131 could image invasive lobular breast cancer, a histologic subtype of breast cancer which is often undetectable by mammography.Conclusions: An HSP90-targeting nIR probe is sensitive and specific in imaging all molecular subtypes of murine and human breast cancer, with higher uptake in aggressive and highly metastatic clones. Clinical studies with Hsp90-targeting nIR probes will be initiated shortly. Clin Cancer Res; 23(24); 7531-42. ©2017 AACR.


Subject(s)
Breast Neoplasms/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Peptides, Cyclic/administration & dosage , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , HSP90 Heat-Shock Proteins/genetics , Humans , Ligands , Mice , Peptides, Cyclic/chemistry , Signal Transduction/drug effects , Spectroscopy, Near-Infrared , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...