Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters

Publication year range
1.
Mol Ther ; 25(3): 580-592, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28187946

ABSTRACT

Immunotherapy with T cells expressing the chimeric antigen receptor (CAR) specific for the CD19 antigen (CD19.CAR-Ts) is a very effective treatment in B cell lymphoid malignancies. However, B cell aplasia and cytokine release syndrome (CRS) secondary to the infusion of CD19.CAR-Ts remain significant drawbacks. The inclusion of safety switches into the vector encoding the CAR is seen as the safest method to terminate the effects of CD19.CAR-Ts in case of severe toxicities or after achieving long-term sustained remissions. By contrast, the complete elimination of CD19.CAR-Ts when CRS occurs may jeopardize clinical responses as CRS and antitumor activity seem to concur. We have demonstrated, in a humanized mouse model, that the inducible caspase-9 (iC9) safety switch can eliminate CD19.CAR-Ts in a dose-dependent manner, allowing either a selective containment of CD19.CAR-T expansion in case of CRS or complete deletion on demand granting normal B cell reconstitution.


Subject(s)
Antigens, CD19/immunology , Caspase 9/metabolism , Cytotoxicity, Immunologic , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Apoptosis/genetics , Apoptosis/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Proliferation , Cell Survival , Gene Expression , Gene Order , Genetic Vectors/genetics , Graft Survival , Hematopoietic Stem Cell Transplantation , Humans , Immunotherapy, Adoptive , Lymphocyte Activation/immunology , Mice , Molecular Imaging , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Protein Binding/immunology , Receptors, Antigen, T-Cell/genetics
2.
Mol Ther ; 24(1): 175-83, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26310629

ABSTRACT

Despite many clinical trials conducted with oncolytic viruses, the exact tumor-level mechanisms affecting therapeutic efficacy have not been established. Currently there are no biomarkers available that would predict the clinical outcome to any oncolytic virus. To assess the baseline immunological phenotype and find potential prognostic biomarkers, we monitored mRNA expression levels in 31 tumor biopsy or fluid samples from 27 patients treated with oncolytic adenovirus. Additionally, protein expression was studied from 19 biopsies using immunohistochemical staining. We found highly significant changes in several signaling pathways and genes associated with immune responses, such as B-cell receptor signaling (P < 0.001), granulocyte macrophage colony-stimulating factor (GM-CSF) signaling (P < 0.001), and leukocyte extravasation signaling (P < 0.001), in patients surviving a shorter time than their controls. In immunohistochemical analysis, markers CD4 and CD163 were significantly elevated (P = 0.020 and P = 0.016 respectively), in patients with shorter than expected survival. Interestingly, T-cell exhaustion marker TIM-3 was also found to be significantly upregulated (P = 0.006) in patients with poor prognosis. Collectively, these data suggest that activation of several functions of the innate immunity before treatment is associated with inferior survival in patients treated with oncolytic adenovirus. Conversely, lack of chronic innate inflammation at baseline may predict improved treatment outcome, as suggested by good overall prognosis.


Subject(s)
Adenoviridae/physiology , Gene Expression Profiling/methods , Immunity, Innate , Neoplasms/genetics , Neoplasms/therapy , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , CD4 Antigens/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasms/immunology , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Prognosis , Receptors, Cell Surface/metabolism , Treatment Outcome
3.
Cancer Immunol Immunother ; 65(6): 741-51, 2016 06.
Article in English | MEDLINE | ID: mdl-27129972

ABSTRACT

Neutrophil elastase (NE) is an innate immune cell-derived inflammatory mediator that we have shown increases the presentation of tumor-associated peptide antigens in breast cancer. In this study, we extend these observations to show that NE uptake has a broad effect on enhancing antigen presentation by breast cancer cells. We show that NE increases human leukocyte antigen (HLA) class I expression on the surface of breast cancer cells in a concentration and time-dependent manner. HLA class I upregulation requires internalization of enzymatically active NE. Western blots of NE-treated breast cancer cells confirm that the expression of total HLA class I as well as the antigen-processing machinery proteins TAP1, LMP2, and calnexin does not change following NE treatment. This suggests that NE does not increase the efficiency of antigen processing; rather, it mediates the upregulation of HLA class I by stabilizing and reducing membrane recycling of HLA class I molecules. Furthermore, the effects of NE extend beyond breast cancer since the uptake of NE by EBV-LCL increases the presentation of HLA class I-restricted viral peptides, as shown by their increased sensitivity to lysis by EBV-specific CD8+ T cells. Together, our results show that NE uptake increases the responsiveness of breast cancer cells to adaptive immunity by broad upregulation of membrane HLA class I and support the conclusion that the innate inflammatory mediator NE enhances tumor cell recognition and increases tumor sensitivity to the host adaptive immune response.


Subject(s)
Antigen Presentation/immunology , Gene Expression Regulation , Histocompatibility Antigens Class I/genetics , Leukocyte Elastase/metabolism , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/immunology , Humans , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
4.
Int J Cancer ; 136(5): 1065-72, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25042001

ABSTRACT

Oncolytic Western Reserve strain vaccinia virus selective for epidermal growth factor receptor pathway mutations and tumor-associated hypermetabolism was armed with human granulocyte-macrophage colony-stimulating factor (GMCSF) and a tdTomato fluorophore. As the assessment of immunological responses to human transgenes is challenging in the most commonly used animal models, we used immunocompetent Syrian golden hamsters, known to be sensitive to human GMCSF and semipermissive to vaccinia virus. Efficacy was initially tested in vitro on various human and hamster cell lines and oncolytic potency of transgene-carrying viruses was similar to unarmed virus. The hGMCSF-encoding virus was able to completely eradicate subcutaneous pancreatic tumors in hamsters, and to fully protect the animals from subsequent rechallenge with the same tumor. Induction of specific antitumor immunity was also shown by ex vivo co-culture experiments with hamster splenocytes. In addition, histological examination revealed increased infiltration of neutrophils and macrophages in GMCSF-virus-treated tumors. These findings help clarify the mechanism of action of GMCSF-armed vaccinia viruses undergoing clinical trials.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Oncolytic Virotherapy , Pancreatic Neoplasms/immunology , Vaccinia virus/genetics , Virus Replication/immunology , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Chlorocebus aethiops , Coculture Techniques , Cricetinae , DNA, Viral/genetics , Humans , Immunoenzyme Techniques , Macrophages , Mesocricetus , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes , Vaccinia virus/immunology , Vero Cells , Xenograft Model Antitumor Assays
5.
Int J Cancer ; 135(3): 720-30, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24374597

ABSTRACT

Sarcomas are a relatively rare cancer, but often incurable at the late metastatic stage. Oncolytic immunotherapy has gained attention over the past years, and a wide range of oncolytic viruses have been delivered via intratumoral injection with positive safety and promising efficacy data. Here, we report preclinical and clinical results from treatment of sarcoma with oncolytic adenovirus Ad5/3-D24-GMCSF (CGTG-102). Ad5/3-D24-GMCSF is a serotype chimeric oncolytic adenovirus coding for human granulocyte-macrophage colony-stimulating factor (GM-CSF). The efficacy of Ad5/3-D24-GMCSF was evaluated on a panel of soft-tissue sarcoma (STS) cell lines and in two animal models. Sarcoma specific human data were also collected from the Advanced Therapy Access Program (ATAP), in preparation for further clinical development. Efficacy was seen in both in vitro and in vivo STS models. Fifteen patients with treatment-refractory STS (13/15) or primary bone sarcoma (2/15) were treated in ATAP, and treatments appeared safe and well-tolerated. A total of 12 radiological RECIST response evaluations were performed, and two cases of minor response, six cases of stable disease and four cases of progressive disease were detected in patients progressing prior to virus treatment. Overall, the median survival time post treatment was 170 days. One patient is still alive at 1,459 days post virus treatment. In summary, Ad5/3-D24-GMCSF appears promising for the treatment of advanced STS; a clinical trial for treatment of refractory injectable solid tumors including STS is ongoing.


Subject(s)
Adenoviridae/genetics , Genetic Therapy , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Oncolytic Virotherapy , Sarcoma/therapy , Animals , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Injections, Intralesional , Mesocricetus , Mice , Mice, Nude , Prognosis , Sarcoma/blood , Sarcoma/mortality , Survival Rate , Tumor Cells, Cultured , Virus Replication , Xenograft Model Antitumor Assays
6.
Mol Ther ; 21(6): 1212-23, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23546299

ABSTRACT

Oncolytic adenoviruses and certain chemotherapeutics can induce autophagy and immunogenic cancer cell death. We hypothesized that the combination of oncolytic adenovirus with low-dose temozolomide (TMZ) is safe, effective, and capable of inducing antitumor immune responses. Metronomic low-dose cyclophosphamide (CP) was added to selectively reduce regulatory T-cells. Preclinically, combination therapy inhibited tumor growth, increased autophagy, and triggered immunogenic cell death as indicated by elevated calreticulin, adenosine triphosphate (ATP) release, and nuclear protein high-mobility group box-1 (HMGB1) secretion. A total of 41 combination treatments given to 17 chemotherapy-refractory cancer patients were well tolerated. We observed anti- and proinflammatory cytokine release, evidence of virus replication, and induction of neutralizing antibodies. Tumor cells showed increased autophagy post-treatment. Release of HMGB1 into serum--a possible indicator of immune response--increased in 60% of treatments, and seemed to correlate with tumor-specific T-cell responses, observed in 10/15 cases overall (P = 0.0833). Evidence of antitumor efficacy was seen in 67% of evaluable treatments with a trend for increased survival over matched controls treated with virus only. In summary, the combination of oncolytic adenovirus with low-dose TMZ and metronomic CP increased tumor cell autophagy, elicited antitumor immune responses, and showed promising safety and efficacy.


Subject(s)
Adenoviridae/genetics , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Dacarbazine/analogs & derivatives , Neoplasms/therapy , Oncolytic Virotherapy/methods , Adenosine Triphosphate/metabolism , Adenoviridae/physiology , Adolescent , Adult , Aged , Animals , Antibodies, Neutralizing/blood , Calreticulin/metabolism , Cell Death/drug effects , Cell Line, Tumor , Child , Combined Modality Therapy/methods , Cyclophosphamide/pharmacology , Cytokines/blood , DNA, Viral/blood , Dacarbazine/pharmacology , Dose-Response Relationship, Drug , Female , HMGB1 Protein/blood , HMGB1 Protein/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Microscopy, Electron , Middle Aged , Oncolytic Viruses/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Temozolomide , Virus Replication , Xenograft Model Antitumor Assays , Young Adult
7.
Mol Ther ; 20(11): 2076-86, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22828500

ABSTRACT

Oncolytic viruses represent a multifaceted tool for cancer treatment. In addition to specific killing of cancer cells (oncolysis), these agents also provide danger signals prompting the immune system to stimulate an antitumor immune response. To increase adenovirus adjuvancy, we engineered the genome of Ad5D24 by inserting 18 immunostimulatory islands (Ad5D24-CpG). The toxicity and immunogenicity profile of Ad5D24-CpG showed that the safety of the maternal virus was retained. The efficacy of the CpG-enriched virus was assessed in a xenograft model of lung cancer where a significant increase in antitumor effect was seen in comparison with controls. When the experiment was repeated in animal depleted of natural killer (NK) cells, Ad5D24-CpG lost its advantage. The same was seen when Toll-like receptor (TLR)9 was blocked systemically. In a syngeneic model of melanoma (B16-OVA), we observed a significant increase of OVA-specific T cells and a decrease of activation of myeloid-derived suppressor cells in Ad5D24-CpG-treated mice. In conclusion, we have generated the first genetically modified oncolytic adenovirus backbone able to enhance TLR9-stimulation for increased antitumor activity.


Subject(s)
Adenoviridae/genetics , Lung Neoplasms/therapy , Melanoma/therapy , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Toll-Like Receptor 9/agonists , Adenoviridae/immunology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival , Combined Modality Therapy , CpG Islands/immunology , HEK293 Cells , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , Myeloid Cells , NF-kappa B/metabolism , Oligodeoxyribonucleotides/pharmacology , Oligodeoxyribonucleotides/therapeutic use , Oncolytic Viruses/immunology , Toll-Like Receptor 9/metabolism , Xenograft Model Antitumor Assays
8.
Mol Ther ; 20(9): 1821-30, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22871667

ABSTRACT

Twenty-five patients with chemotherapy refractory cancer were treated with a fully serotype 3-based oncolytic adenovirus Ad3-hTERT-E1A. In mice, Ad3 induced higher amounts of cytokines but less liver damage than Ad5 or Ad5/3. In humans, the only grade 3 adverse reactions were self-limiting cytopenias and generally the safety profile resembled Ad5-based oncolytic viruses. Patients that had been previously treated with Ad5 viruses presented longer lasting lymphocytopenia but no median increase in Ad3-specific T-cells in blood, suggesting immunological activity against antigens other than Ad3 hexon. Frequent alterations in antitumor T-cells in blood were seen regardless of previous virus exposure. Neutralizing antibodies against Ad3 increased in all patients, whereas Ad5 neutralizing antibodies remained stable. Treatment with Ad3-hTERT-E1A resulted in re-emergence of Ad5 viruses from previous treatments into blood and vice versa. Signs of possible efficacy were seen in 11/15 (73%) patients evaluable for tumor markers, four of which were treated only intravenously. Particularly promising results were seen in breast cancer patients and especially those receiving concomitant trastuzumab. Taken together, Ad3-hTERT-E1A seems safe for further clinical testing or development of armed versions. It offers an immunologically attractive alternative, with possible pharmacodynamic differences and a different receptor compared to Ad5.


Subject(s)
Adenoviridae/immunology , Genetic Therapy , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Adenoviridae/genetics , Adult , Aged , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/therapy , Drug Resistance, Neoplasm , Female , Genes, Viral , Genetic Vectors , Humans , Male , Mice , Middle Aged , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/mortality , Oncolytic Viruses/genetics , Survival Rate , T-Lymphocytes/immunology , Trastuzumab , Virus Replication
9.
Int J Cancer ; 130(8): 1937-47, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-21630267

ABSTRACT

The safety of oncolytic viruses for treatment of cancer has been shown in clinical trials while antitumor efficacy has often remained modest. As expression of the coxsackie-adenovirus receptor may be variable in advanced tumors, we developed Ad5-D24-RGD, a p16/Rb pathway selective oncolytic adenovirus featuring RGD-4C modification of the fiber. This allows viral entry through alpha-v-beta integrins frequently highly expressed in advanced tumors. Advanced tumors are often immunosuppressive which results in lack of tumor eradication despite abnormal epitopes being present. Granulocyte-macrophage colony stimulating factor (GMCSF) is a potent activator of immune system with established antitumor properties. To stimulate antitumor immunity and break tumor associated immunotolerance, we constructed Ad5-RGD-D24-GMCSF, featuring GMCSF controlled by the adenoviral E3 promoter. Preliminary safety of Ad5-D24-RGD and Ad5-RGD-D24-GMCSF for treatment of human cancer was established. Treatments with Ad5-D24-RGD (N = 9) and Ad5-RGD-D24-GMCSF (N = 7) were well tolerated. Typical side effects were grade 1-2 fatigue, fever and injection site pain. 77% (10/13) of evaluable patients showed virus in circulation for at least 2 weeks. In 3 out of 6 evaluable patients, disease previously progressing stabilized after a single treatment with Ad5-RGD-D24-GMCSF. In addition, 2/3 patients had stabilization or reduction in tumor marker levels. All patients treated with Ad5-D24-RGD showed disease progression in radiological analysis, although 3/6 had temporary reduction or stabilization of marker levels. Induction of tumor and adenovirus specific immunity was demonstrated with ELISPOT in Ad5-RGD-D24-GMCSF treated patients. RGD modified oncolytic adenoviruses with or without GMCSF seem safe for further clinical development.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Neoplasms/therapy , Oligopeptides/metabolism , Oncolytic Virotherapy/methods , Adenoviridae/genetics , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , DNA, Viral/genetics , Drug Resistance, Neoplasm , Fatigue/etiology , Female , Fever/etiology , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Integrins/metabolism , Male , Middle Aged , Neoplasms/metabolism , Neoplasms/virology , Oligopeptides/genetics , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/genetics , Real-Time Polymerase Chain Reaction , Treatment Outcome , Viral Load , Virus Replication/genetics
10.
Mol Ther ; 19(10): 1858-66, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21792178

ABSTRACT

Oncolytic adenoviruses are an emerging experimental approach for treatment of tumors refractory to available modalities. Although preclinical results have been promising, and clinical safety has been excellent, it is also apparent that tumors can become virus resistant. The resistance mechanisms acquired by advanced tumors against conventional therapies are increasingly well understood, which has allowed development of countermeasures. To study this in the context of oncolytic adenovirus, we developed two in vivo models of acquired resistance, where initially sensitive tumors eventually gain resistance and relapse. These models were used to investigate the phenomenon on RNA and protein levels using two types of analysis of microarray data, quantitative reverse transcriptase-polymerase chain reaction and immunohistochemistry. Interferon (IFN) signaling pathways were found upregulated and Myxovirus resistance protein A (MxA) expression was identified as a marker correlating with resistance, while transplantation experiments suggested a role for tumor stroma in maintaining resistance. Furthermore, pathway analysis suggested potential therapeutic targets in oncolytic adenovirus-resistant cells. Improved understanding of the antiviral phenotype causing tumor recurrence is of key importance in order to improve treatment of advanced tumors with oncolytic adenoviruses. Given the similarities between mechanisms of action, this finding might be relevant for other oncolytic viruses as well.


Subject(s)
Adenoviridae/physiology , Interferons/biosynthesis , Oncolytic Virotherapy , Animals , Base Sequence , Cell Line, Tumor , DNA Primers , Female , Humans , Immunohistochemistry , Mice , Mice, SCID , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
11.
Mol Ther ; 19(9): 1737-46, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21673660

ABSTRACT

Patients with advanced solid tumors refractory to and progressing after conventional therapies were treated with three different regimens of low-dose cyclophosphamide (CP) in combination with oncolytic adenovirus. CP was given with oral metronomic dosing (50 mg/day, N = 21), intravenously (single 1,000 mg dose, N = 7) or both (N = 7). Virus was injected intratumorally. Controls (N = 8) received virus without CP. Treatments were well tolerated and safe regardless of schedule. Antibody formation and virus replication were not affected by CP. Metronomic CP (oral and oral + intravenous schedules) decreased regulatory T cells (T(regs)) without compromising induction of antitumor or antiviral T-cell responses. Oncolytic adenovirus given together with metronomic CP increased cytotoxic T cells and induced Th1 type immunity on a systemic level in most patients. All CP regimens resulted in higher rates of disease control than virus only (all P < 0.0001) and the best progression-free (PFS) and overall survival (OS) was seen in the oral + intravenous group. One year PFS and OS were 53 and 42% (P = 0.0016 and P < 0.02 versus virus only), respectively, both which are unusually high for chemotherapy refractory patients. We conclude that low-dose CP results in immunological effects appealing for oncolytic virotherapy. While these first-in-human data suggest good safety, intriguing efficacy and extended survival, the results should be confirmed in a randomized trial.


Subject(s)
Antineoplastic Agents/administration & dosage , Cyclophosphamide/administration & dosage , Neoplasms/drug therapy , Oncolytic Virotherapy/methods , T-Lymphocytes, Regulatory/immunology , Adenoviridae/genetics , Adolescent , Adult , Aged , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Child , Combined Modality Therapy , Cricetinae , Cyclophosphamide/immunology , Cyclophosphamide/therapeutic use , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Genetic Vectors , Humans , Male , Mesocricetus , Middle Aged , Neoplasms/immunology , Treatment Outcome , Young Adult
12.
J Pers Med ; 12(11)2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36422104

ABSTRACT

(1) Background: The outbreak of the COVID-19 pandemic represented a real challenge for all of humanity. Characterized by a complex spectrum of signs and symptoms, by various severity degrees, the disease spread rapidly around the world. After more than two and half years since the beginning of COVID-19 pandemic, in the context of a paradoxical, enigmatic, and relentless COVID-19, the objective of the current study was to evaluate the characteristics and evolution of patients with SARS-CoV-2 infection, hospitalized in "St. Parascheva" Clinical Hospital of Infectious Diseases (Iasi, Romania). (2) Methods: This is a retrospective study that used the medical database recorded between July and November 2021 in order to highlight the characteristics of SARS-CoV-2 infection in patients from the northeastern region of Romania. (3) Results: We enrolled in the study a total of 1732 SARS-CoV-2 infected patients, mean age 67 ± 3.4 years, the female gender predominating (987 cases; 56.98%) as well as patients from the urban environment (982 patients; 56.69%). Moderate form of the disease predominated (814 cases; 47%), pulmonary imaging changes were found in 1042 (60.16%) cases, and 1242 (71.71%) patients had at least one underlying disease. After a median length of hospitalization of 9.5 days, 1359 (78.46%) patients were discharged cured, 48 (2.77%) were transferred to other services by decompensating the associated pathologies, 302 (17.43%) patients needed extensive support in the intensive care unit and there were 325 (18.76%) deaths. (4) Conclusions: The epidemiological characteristics of SARS-CoV-2 infection recorded in our study were mostly the same as characteristics of COVID-19 from all over the world.

13.
J Virol ; 84(2): 856-66, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19906926

ABSTRACT

Oncolytic vaccinia viruses have shown compelling results in preclinical cancer models and promising preliminary safety and antitumor activity in early clinical trials. However, to facilitate systemic application it would be useful to improve tumor targeting and antitumor efficacy further. Here we report the generation of vvdd-VEGFR-1-Ig, a targeted and armed oncolytic vaccinia virus. Tumor targeting was achieved by deletion of genes for thymidine kinase and vaccinia virus growth factor, which are necessary for replication in normal but not in cancer cells. Given the high vascularization typical of kidney cancers, we armed the virus with the soluble vascular endothelial growth factor (VEGF) receptor 1 protein for an antiangiogenic effect. Systemic application of high doses of vvdd-VEGFR-1-Ig resulted in cytokine induction in an immunocompromised mouse model. Upon histopathological analysis, splenic extramedullary hematopoiesis was seen in all virus-injected mice and was more pronounced in the vvdd-VEGFR-1-Ig group. Analysis of the innate immune response after intravenous virus injection revealed high transient and dose-dependent cytokine elevations. When medium and low doses were used for intratumoral or intravenous injection, vvdd-VEGFR-1-Ig exhibited a stronger antitumor effect than the unarmed control. Furthermore, expression of VEGFR-1-Ig was confirmed, and a concurrent antiangiogenic effect was seen. In an immunocompetent model, systemic vvdd-VEGFR-1-Ig exhibited superior antitumor efficacy compared to the unarmed control virus. In conclusion, the targeted and armed vvdd-VEGFR-1-Ig has promising anticancer activity in renal cell cancer models. Extramedullary hematopoiesis may be a sensitive indicator of vaccinia virus effects in mice.


Subject(s)
Angiogenesis Inhibitors , Kidney Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses , Vaccinia virus , Vascular Endothelial Growth Factor Receptor-1 , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Immunocompetence , Immunocompromised Host , Kidney/cytology , Kidney/virology , Kidney Neoplasms/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Treatment Outcome , Vaccinia virus/genetics , Vaccinia virus/metabolism , Vascular Endothelial Growth Factor Receptor-1/administration & dosage , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
14.
Mol Ther ; 18(10): 1874-84, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20664527

ABSTRACT

Augmenting antitumor immunity is a promising way to enhance the potency of oncolytic adenoviral therapy. Granulocyte-macrophage colony-stimulating factor (GMCSF) can mediate antitumor effects by recruiting natural killer cells and by induction of tumor-specific CD8(+) cytotoxic T-lymphocytes. Serotype 5 adenoviruses (Ad5) are commonly used in cancer gene therapy. However, expression of the coxsackie-adenovirus receptor is variable in many advanced tumors and preclinical data have demonstrated an advantage for replacing the Ad5 knob with the Ad3 knob. Here, a 5/3 capsid chimeric and p16-Rb pathway selective oncolytic adenovirus coding for GMCSF was engineered and tested preclinically. A total of 21 patients with advanced solid tumors refractory to standard therapies were then treated intratumorally and intravenously with Ad5/3-D24-GMCSF, which was combined with low-dose metronomic cyclophosphamide to reduce regulatory T cells. No severe adverse events occurred. Analysis of pretreatment samples of malignant pleural effusion and ascites confirmed the efficacy of Ad5/3-D24-GMCSF in transduction and cell killing. Evidence of biological activity of the virus was seen in 13/21 patients and 8/12 showed objective clinical benefit as evaluated by radiology with Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Antiadenoviral and antitumoral immune responses were elicited after treatment. Thus, Ad5/3-D24-GMCSF seems safe in treating cancer patients and promising signs of efficacy were seen.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Neoplasms/therapy , Oncolytic Virotherapy/methods , Adolescent , Adult , Aged , Animals , Cell Line , Cell Line, Tumor , Cricetinae , Cyclophosphamide/therapeutic use , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Immunosuppressive Agents/therapeutic use , Male , Mesocricetus , Middle Aged , Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Young Adult
15.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33771892

ABSTRACT

BACKGROUND: The cancer-testis antigen MAGE-A4 is an attractive target for T-cell-based immunotherapy, especially for indications with unmet clinical need like non-small cell lung or triple-negative breast cancer. METHODS: An unbiased CD137-based sorting approach was first used to identify an immunogenic MAGE-A4-derived epitope (GVYDGREHTV) that was properly processed and presented on human leukocyte antigen (HLA)-A2 molecules encoded by the HLA-A*02:01 allele. To isolate high-avidity T cells via subsequent multimer sorting, an in vitro priming approach using HLA-A2-negative donors was conducted to bypass central tolerance to this self-antigen. Pre-clinical parameters of safety and activity were assessed in a comprehensive set of in vitro and in vivo studies. RESULTS: A MAGE-A4-reactive, HLA-A2-restricted T-cell receptor (TCR) was isolated from primed T cells of an HLA-A2-negative donor. The respective TCR-T-cell (TCR-T) product bbT485 was demonstrated pre-clinically to have a favorable safety profile and superior in vivo potency compared with TCR-Ts expressing a TCR derived from a tolerized T-cell repertoire to self-antigens. This natural high-avidity TCR was found to be CD8 co-receptor independent, allowing effector functions to be elicited in transgenic CD4+ T helper cells. These CD4+ TCR-Ts supported an anti-tumor response by direct killing of MAGE-A4-positive tumor cells and upregulated hallmarks associated with helper function, such as CD154 expression and release of key cytokines on tumor-specific stimulation. CONCLUSION: The extensive pre-clinical assessment of safety and in vivo potency of bbT485 provide the basis for its use in TCR-T immunotherapy studies. The ability of this non-mutated high-avidity, co-receptor-independent TCR to activate CD8+ and CD4+ T cells could potentially provide enhanced cellular responses in the clinical setting through the induction of functionally diverse T-cell subsets that goes beyond what is currently tested in the clinic.


Subject(s)
Antigens, Neoplasm/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/transplantation , Immunotherapy, Adoptive , Neoplasm Proteins/immunology , Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , A549 Cells , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , CD8 Antigens/genetics , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Coculture Techniques , Cytotoxicity, Immunologic , Female , HEK293 Cells , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Humans , Immunodominant Epitopes , K562 Cells , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Phenotype , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Tumor Burden , Xenograft Model Antitumor Assays
16.
J Gene Med ; 12(5): 435-45, 2010 May.
Article in English | MEDLINE | ID: mdl-20440754

ABSTRACT

BACKGROUND: Adenoviruses can cause severe toxicity in children and in immunocompromised adults, and therefore a means to abrogate replication would be useful. With regard to cancer treatment, replication competent oncolytic adenoviruses have been safe in humans, although their efficacy has been variable. Therefore, more effective agents are now entering clinical testing and, consequently, replication-associated side effects remain a concern. Preclinical analysis of replication related toxicity has been hampered by a lack of permissive models. Therefore, it has been difficult to study modulation of human adenovirus replication in immune competent animals. METHODS: We investigated four different hamster carcinoma cell lines for transduction and cell killing potency in vitro and in vivo. Gene transfer was assessed using replication-deficient adenoviruses expressing luciferase. Cell killing was studied in vitro and in vivo using an oncolytic adenovirus that kills tumor cells by viral replication. After the most promising animal model had been selected, abrogation of virus replication was assessed in vitro and in vivo using a TCID(50) assay. RESULTS: The results obtained suggest wild-type adenovirus replication in all four tested Syrian hamster cell lines and also normal organs. Virus replication could be abrogated with chlorpromazine, cidofovir and cytosine arabinoside, and the effect occurred subsequent to nuclear delivery of the viral genome. Attenuation of virus replication also was seen in vivo both in tumors and the liver. CONCLUSIONS: Syrian hamsters may comprise a valuable immune competent model for evaluating anti-adenoviral drugs. Furthermore, chlorpromazine or cidofovir might be useful in case of adenovirus replication-associated symptoms in humans.


Subject(s)
Adenoviruses, Human/drug effects , Adenoviruses, Human/physiology , Chlorpromazine/pharmacology , Cytosine/analogs & derivatives , Immunocompetence/immunology , Mesocricetus/virology , Organophosphonates/pharmacology , Virus Replication/drug effects , Adenoviridae Infections/virology , Animals , Biological Transport/drug effects , Cell Death/drug effects , Cell Line , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/virology , Cidofovir , Cricetinae , Cytarabine/pharmacology , Cytosine/pharmacology , Humans , Immunocompetence/drug effects , Mesocricetus/immunology , Neoplasms/pathology , Neoplasms/virology , Organ Specificity/drug effects , Transduction, Genetic
17.
J Transl Med ; 8: 80, 2010 Aug 23.
Article in English | MEDLINE | ID: mdl-20727221

ABSTRACT

BACKGROUND: Colorectal cancer is often a deadly disease and cannot be cured at metastatic stage. Oncolytic adenoviruses have been considered as a new therapeutic option for treatment of refractory disseminated cancers, including colorectal cancer. The safety data has been excellent but tumor transduction and antitumor efficacy especially in systemic administration needs to be improved. METHODS: Here, the utility of αvß integrin targeting moiety Arg-Gly-Asp (RGD) in the Lys-Lys-Thr-Lys (KKTK) domain of the fiber shaft or in the HI-loop of adenovirus serotype 5 for increased tumor targeting and antitumor efficacy was evaluated. To this end, novel spleen-to-liver metastatic colorectal cancer mouse model was used and the antitumor efficacy was evaluated with magnetic resonance imaging (MRI). RESULTS: Both modifications (RGD in the HI-loop or in the fiber shaft) increased gene transfer efficacy in colorectal cancer cell lines and improved tumor-to-normal ratio in systemic administration of the vector. CONCLUSIONS: Antitumor potency was not compromised with RGD modified viruses suggesting increased safety profile and tumor specificity.


Subject(s)
Adenoviridae/chemistry , Adenoviridae/genetics , Colorectal Neoplasms/therapy , Magnetic Resonance Imaging , Oligopeptides/metabolism , Receptors, Vitronectin/metabolism , Adenoviridae/physiology , Animals , Capsid/metabolism , Cell Line, Tumor , Colorectal Neoplasms/pathology , Cytotoxicity, Immunologic , Gene Transfer Techniques , Genetic Vectors/pharmacokinetics , Humans , Liver Neoplasms/secondary , Mice , Neoplasm Metastasis , Organ Specificity , Splenic Neoplasms/secondary , Tissue Distribution , Virus Replication , Xenograft Model Antitumor Assays
18.
Clin Cancer Res ; 25(24): 7340-7350, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31558475

ABSTRACT

PURPOSE: Current protocols for CD19 chimeric antigen receptor-expressing T cells (CD19.CAR-T cells) require recipients to tolerate preinfusion cytoreductive chemotherapy, and the presence of sufficient target antigen on normal or malignant B cells. PATIENTS AND METHODS: We investigated whether additional stimulation of CD19.CAR-T cells through their native receptors can substitute for cytoreductive chemotherapy, inducing expansion and functional persistence of CD19.CAR-T even in patients in remission of B-cell acute lymphocytic leukemia. We infused a low dose of CD19.CAR-modified virus-specific T cells (CD19.CAR-VST) without prior cytoreductive chemotherapy into 8 patients after allogeneic stem cell transplant. RESULTS: Absent virus reactivation, we saw no CD19.CAR-VST expansion. In contrast, in patients with viral reactivation, up to 30,000-fold expansion of CD19.CAR-VSTs was observed, with depletion of CD19+ B cells. Five patients remain in remission at 42-60+ months. CONCLUSIONS: Dual T-cell receptor and CAR stimulation can thus potentiate effector cell expansion and CAR-target cell killing, even when infusing low numbers of effector cells without cytoreduction.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive/methods , Lymphoma, B-Cell/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/transplantation , Adenoviridae/physiology , Adolescent , Antigens, CD19/metabolism , Child , Child, Preschool , Genetic Vectors , Herpesvirus 4, Human/physiology , Humans , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/virology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/virology , Retroviridae/physiology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Young Adult
19.
Immunol Res ; 36(1-3): 229-36, 2006.
Article in English | MEDLINE | ID: mdl-17337783

ABSTRACT

Endometriosis is a chronic, debilitating disease, associated with pelvic pain and infertility. Recent epidemiological studies suggest that women with endometriosis are at increased risk for ovarian cancer. Although the causative factors for both endometriosis and ovarian cancer remain largely unknown, several similarities between the proposed etiology of ovarian cancer and the observed pathophysiology of endometriosis have been reported. MUC1 glycoprotein is present in endometriotic lesions and overexpressed in epithelial ovarian tumors. We are currently studying immunity to MUC1 antigen in newly emerging preclinical models for endometriosis and ovarian cancer and exploring the potential for immune therapy/prevention with MUC1 in both diseases.


Subject(s)
Cancer Vaccines , Endometriosis/etiology , Mucin-1/immunology , Ovarian Neoplasms/complications , Ovarian Neoplasms/prevention & control , Animals , Endometriosis/metabolism , Female , Humans , Mucin-1/metabolism , Ovarian Neoplasms/metabolism , Precancerous Conditions/metabolism , Risk Factors
20.
Oncotarget ; 6(31): 30675-703, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26362400

ABSTRACT

The emergence of genetic engineering at the beginning of the 1970's opened the era of biomedical technologies, which aims to improve human health using genetic manipulation techniques in a clinical context. Gene therapy represents an innovating and appealing strategy for treatment of human diseases, which utilizes vehicles or vectors for delivering therapeutic genes into the patients' body. However, a few past unsuccessful events that negatively marked the beginning of gene therapy resulted in the need for further studies regarding the design and biology of gene therapy vectors, so that this innovating treatment approach can successfully move from bench to bedside. In this paper, we review the major gene delivery vectors and recent improvements made in their design meant to overcome the issues that commonly arise with the use of gene therapy vectors. At the end of the manuscript, we summarized the main advantages and disadvantages of common gene therapy vectors and we discuss possible future directions for potential therapeutic vectors.


Subject(s)
Gene Transfer Techniques , Genetic Engineering/methods , Genetic Therapy/methods , Genetic Vectors/genetics , Adenoviridae/genetics , Dependovirus/genetics , Humans , Lentivirus/genetics , Plasmids/genetics , RNA Interference , RNA, Small Interfering/genetics , Retroviridae/genetics , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL