Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Mol Ther ; 21(6): 1212-23, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23546299

RESUMEN

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.


Asunto(s)
Adenoviridae/genética , Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Dacarbazina/análogos & derivados , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adenosina Trifosfato/metabolismo , Adenoviridae/fisiología , Adolescente , Adulto , Anciano , Animales , Anticuerpos Neutralizantes/sangre , Calreticulina/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Niño , Terapia Combinada/métodos , Ciclofosfamida/farmacología , Citocinas/sangre , ADN Viral/sangre , Dacarbazina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Proteína HMGB1/sangre , Proteína HMGB1/metabolismo , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Microscopía Electrónica , Persona de Mediana Edad , Virus Oncolíticos/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Temozolomida , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
2.
Clin Cancer Res ; 19(10): 2734-44, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23493351

RESUMEN

PURPOSE: Multiple injections of oncolytic adenovirus could enhance immunologic response. In the first part of this article, the focus was on immunologic aspects. Sixty patients previously naïve to oncolytic virus and who had white blood cells available were treated. Thirty-nine of 60 were assessed after a single virus administration, whereas 21 of 60 received a "serial treatment" consisting of three injections within 10 weeks. In the second part, we focused on 115 patients treated with a granulocyte macrophage colony-stimulating factor (GM-CSF)-coding capsid chimeric adenovirus, CGTG-102. RESULTS: Following serial treatment, both increase and decrease in antitumor T cells in blood were seen more frequently, findings which are compatible with induction of T-cell immunity and trafficking of T cells to tumors, respectively. Safety was good in both groups. In 115 patients treated with CGTG-102 (Ad5/3-D24-GMCSF), median overall survival was 111 days following single and 277 days after serial treatment in nonrandomized comparison. Switching the virus capsid for avoiding neutralizing antibodies in a serial treatment featuring three different viruses did not impact safety or efficacy. A correlation between antiviral and antitumor T cells was seen (P = 0.001), suggesting that viral oncolysis can result in epitope spreading and breaking of tumor-associated immunologic tolerance. Alternatively, some patients may be more susceptible to induction of T-cell immunity and/or trafficking. CONCLUSIONS: These results provide the first human data linking antiviral immunity with antitumor immunity, implying that oncolytic viruses could have an important role in cancer immunotherapy.


Asunto(s)
Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Linfocitos T/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Adolescente , Adulto , Anciano , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Niño , Anergia Clonal/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-10/inmunología , Interleucina-10/metabolismo , Masculino , Persona de Mediana Edad , Neoplasias/clasificación , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Análisis de Supervivencia , Linfocitos T/metabolismo , Factores de Tiempo , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
3.
Mol Ther ; 20(9): 1821-30, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22871667

RESUMEN

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.


Asunto(s)
Adenoviridae/inmunología , Terapia Genética , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Adenoviridae/genética , Adulto , Anciano , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/terapia , Resistencia a Antineoplásicos , Femenino , Genes Virales , Vectores Genéticos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/mortalidad , Virus Oncolíticos/genética , Tasa de Supervivencia , Linfocitos T/inmunología , Trastuzumab , Replicación Viral
4.
Cancer Res ; 72(7): 1621-31, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22323527

RESUMEN

The immunosuppressive environment of advanced tumors is a primary obstacle to the efficacy of immunostimulatory and vaccine approaches. Here, we report an approach to arm an oncolytic virus with CD40 ligand (CD40L) to stimulate beneficial immunologic responses in patients. A double-targeted chimeric adenovirus controlled by the hTERT promoter and expressing CD40L (CGTG-401) was constructed and nine patients with progressing advanced solid tumors refractory to standard therapies were treated intratumorally. No serious adverse events resulting in patient hospitalization occurred. Moderate or no increases in neutralizing antibodies were seen, suggesting effective Th1 immunologic effects. An assessment of the blood levels of virus indicated 17.5% of the samples (n = 40) were positive at a low level early after treatment, but not thereafter. In contrast, high levels of virus, CD40L, and RANTES were documented locally at the tumor. Peripheral blood mononuclear cells were analyzed by IFN-γ ELISPOT analysis and induction of both survivin-specific and adenovirus-specific T cells was seen. Antitumor T-cell responses were even more pronounced when assessed by intracellular cytokine staining after stimulation with tumor type-specific peptide pools. Of the evaluable patients, 83% displayed disease control at 3 months and in both cases in which treatment was continued the effect was sustained for at least 8 months. Injected and noninjected lesions responded identically. Together, these findings support further clinical evaluation of CGTG-401.


Asunto(s)
Adenoviridae/genética , Ligando de CD40/genética , Inmunoterapia , Neoplasias/terapia , Viroterapia Oncolítica , Adenoviridae/inmunología , Adulto , Anticuerpos Antivirales/análisis , Ligando de CD40/análisis , Quimiocina CCL5/análisis , Femenino , Humanos , Interferón gamma/biosíntesis , Masculino , Persona de Mediana Edad , Imagen Multimodal , Neoplasias/inmunología , Viroterapia Oncolítica/efectos adversos , Tomografía de Emisión de Positrones , Telomerasa/genética , Células TH1/inmunología , Tomografía Computarizada por Rayos X , Virión/aislamiento & purificación
5.
Mol Ther ; 20(1): 221-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22044933

RESUMEN

Calcium channel blockers including verapamil have been proposed to enhance release and antitumor efficacy of oncolytic adenoviruses in preclinical studies but this has not been studied in humans before. Here, we studied if verapamil leads to increased replication of oncolytic adenovirus in cancer patients, as measured by release of virions from tumor cells into the systemic circulation. The study was conducted as a matched case-control study of advanced cancer patients treated with oncolytic adenoviruses with or without verapamil. We observed that verapamil increased mean virus titers present in blood after treatment (P < 0.05). The frequency or severity of adverse events was not increased, nor were cytokine responses or neutralizing antibody levels different between groups. Signs of possible treatment-related clinical benefits were observed in both groups, but there was no significant difference in responses or survival. Thus, our data suggests that the combination of verapamil with oncolytic adenoviruses is safe and well tolerated. Moreover, verapamil treatment seems to result in higher virus titers in blood, indicating enhanced overall replication in tumors. A randomized trial is needed to confirm these findings and to study if enhanced replication results in benefits to patients.


Asunto(s)
Adenoviridae/genética , Bloqueadores de los Canales de Calcio/uso terapéutico , Vectores Genéticos , Neoplasias/terapia , Virus Oncolíticos/genética , Verapamilo/uso terapéutico , Adenoviridae/inmunología , Adolescente , Adulto , Anciano , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Bloqueadores de los Canales de Calcio/efectos adversos , Terapia Combinada , ADN Viral/sangre , Sinergismo Farmacológico , Femenino , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Humanos , Mediadores de Inflamación/sangre , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias/mortalidad , Neoplasias/patología , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/inmunología , Análisis de Supervivencia , Resultado del Tratamiento , Verapamilo/efectos adversos , Carga Viral/efectos de los fármacos , Adulto Joven
6.
Int J Cancer ; 130(8): 1937-47, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21630267

RESUMEN

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.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Neoplasias/terapia , Oligopéptidos/metabolismo , Viroterapia Oncolítica/métodos , Adenoviridae/genética , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , ADN Viral/genética , Resistencia a Antineoplásicos , Fatiga/etiología , Femenino , Fiebre/etiología , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Integrinas/metabolismo , Masculino , Persona de Mediana Edad , Neoplasias/metabolismo , Neoplasias/virología , Oligopéptidos/genética , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Resultado del Tratamiento , Carga Viral , Replicación Viral/genética
7.
Int J Cancer ; 131(1): 253-63, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21834073

RESUMEN

Conventional cancer treatments often have little impact on the course of advanced pancreatic cancer. Although cancer gene therapy with adenoviruses is a promising developmental approach, the primary receptor is poorly expressed in pancreatic cancers which might compromise efficacy and thus targeting to other receptors could be beneficial. Extended stealth delivery, combination with standard chemotherapy or circumvention of host antiadenoviral immune response might improve efficacy further. In this work, capsid-modified adenoviruses were studied for transduction of cell lines and clinical normal and tumor tissue samples. The respective oncolytic viruses were tested for oncolytic activity in vitro and in vivo. Survival was studied in a peritoneally disseminated pancreas cancer model, with or without concurrent gemcitabine while silica implants were utilized for extended intraperitoneal virus delivery. Immunocompetent mice and Syrian hamsters were used to study the effect of silica mediated delivery on antiviral immune responses and subsequent in vivo gene delivery. Capsid modifications selectively enhanced gene transfer to malignant pancreatic cancer cell lines and clinical samples. The respective oncolytic viruses resulted in increased cell killing in vitro, which translated into a survival benefit in mice. Early proinfammatory cytokine responses and formation of antiviral neutralizing antibodies was partially avoided with silica implants. The implant also shielded the virus from pre-existing neutralizing antibodies, while increasing the pancreas/liver gene delivery ratio six-fold. In conclusion, capsid modified adenoviruses would be useful for testing in pancreatic cancer trials. Silica implants might increase the safety and efficacy of the approach.


Asunto(s)
Adenovirus Humanos , Desoxicitidina/análogos & derivados , Viroterapia Oncolítica/métodos , Virus Oncolíticos , Neoplasias Pancreáticas/terapia , Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Animales , Anticuerpos Neutralizantes , Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular Tumoral , Terapia Combinada , Cricetinae , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Femenino , Humanos , Ratones , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/inmunología , Gel de Sílice , Transfección , Gemcitabina
8.
Mol Ther ; 19(9): 1737-46, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21673660

RESUMEN

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.


Asunto(s)
Antineoplásicos/administración & dosificación , Ciclofosfamida/administración & dosificación , Neoplasias/tratamiento farmacológico , Viroterapia Oncolítica/métodos , Linfocitos T Reguladores/inmunología , Adenoviridae/genética , Adolescente , Adulto , Anciano , Animales , Antineoplásicos/inmunología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Niño , Terapia Combinada , Cricetinae , Ciclofosfamida/inmunología , Ciclofosfamida/uso terapéutico , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Vectores Genéticos , Humanos , Masculino , Mesocricetus , Persona de Mediana Edad , Neoplasias/inmunología , Resultado del Tratamiento , Adulto Joven
9.
J Gene Med ; 13(5): 253-61, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21520358

RESUMEN

BACKGROUND: Oncolytic adenoviruses are an attractive strategy for treating cancers resistant to conventional treatments. However, their systemic utility could be limited as a result of the high prevalence of pre-existing immunity towards the vector. Furthermore, neutralizing antibodies (NAbs) may prevent successful intravenous readministration of the same agent. Previous preclinical reports indicate that the NAb response can be partially overcome by modifying the adenoviral fiber knob. However, to date, this strategy has not been evaluated in human patients. METHODS: Twenty-four human patients with advanced cancer were treated with two cycles of oncolytic adenoviruses, featuring three capsid variants: unmodified adenovirus serotype 5 (Ad5), serotype 5 with RGD motif in the HI-loop of the fiber knob (Ad5-RGD) and serotype 5 carrying fiber knob from serotype 3 (Ad5/3). A virus with different fiber structure was used for the second round of treatment and patient serum was analyzed for a neutralizing effect. RESULTS: All patients developed NAbs against the virus that they were treated with. However, the magnitude and velocity of the response varied considerably. When measured just before the second treatment cycle, a differential in serum NAb titer against the first versus second virus was seen in 83% of cases, suggesting that even minor changes in the fiber knob can circumvent neutralization in cancer patients. No correlation between NAb titers and outcome variables was observed. CONCLUSIONS: The results obtained in the present study extend previous preclinical reports into human cancer patients and suggest that modification of the fiber knob is a feasible strategy for circumventing the NAb response in patients receiving multiple rounds of oncolytic adenoviruses.


Asunto(s)
Adenoviridae/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Proteínas de la Cápside/inmunología , Vectores Genéticos/inmunología , Neoplasias , Viroterapia Oncolítica , Adenoviridae/genética , Adolescente , Adulto , Anciano , Terapia Genética , Humanos , Persona de Mediana Edad , Neoplasias/inmunología , Neoplasias/terapia , Resultado del Tratamiento , Adulto Joven
11.
Ann Med ; 43(2): 151-63, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21261555

RESUMEN

BACKGROUND: Successful tumor targeting of systemically administered oncolytic adenoviruses may be hindered by interactions with blood components. MATERIALS AND METHODS: Blood distribution of oncolytic adenoviruses featuring type 5 adenovirus fiber, 5/3 capsid chimerism, or RGD-4C in the fiber knob was investigated in vitro and in patients with refractory solid tumors. RESULTS: Virus titers and prevalence in serum of patients increased over the first post-treatment week, suggesting replication. Detection of low virus loads was more sensitive in blood clots than in serum, although viral levels > 500 viral particles/mL did not differ significantly between both sample types. While adenovirus bound to erythrocytes, platelets, granulocytes, and peripheral blood mononuclear cells in vitro, the virus was mainly detectable in erythrocytes and granulocytes in cancer patients. Taken together with a temporary post-treatment decrease in thrombocyte counts, platelet activation by adenovirus and subsequent clearance seem likely to occur in humans. Fiber modifications had limited observed effect on virus distribution in blood cell compartments. Neutrophils, monocytes and cytotoxic T lymphocytes were the major leukocyte subpopulations interacting with adenoviruses. CONCLUSION: Serum and blood clots are relevant to estimate oncolytic adenovirus replication. Insight into viral interactions with blood cells may contribute to the development of new strategies for tumor delivery.


Asunto(s)
Adenoviridae/metabolismo , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Coagulación Sanguínea , Plaquetas/metabolismo , Eritrocitos/metabolismo , Granulocitos/metabolismo , Humanos , Monocitos/metabolismo , Neoplasias/patología , Neutrófilos/metabolismo , Virus Oncolíticos/metabolismo , Linfocitos T Citotóxicos/metabolismo , Replicación Viral
12.
Mol Pharm ; 8(1): 12-28, 2011 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-21126047

RESUMEN

Although results of cancer treatment have improved steadily, metastatic solid tumors can be cured only rarely and therefore new modalities are needed. Tumors often become apoptosis-resistant and capable of excluding drugs during therapy. Similar mechanisms of resistance apply to many treatment regimens, and cross-resistance between different chemotherapeutics often limits the treatment options. Therefore, loss of efficacy may occur simultaneously for different chemotherapeutics. One experimental strategy with an increasing amount of clinical evidence is oncolytic viruses, which replicate preferentially in tumor cells by taking advantage of cancer-specific cellular changes. Adenoviruses are the most widely clinically used oncolytic agents. Replication of oncolytic virus per se kills tumor cells, but oncolysis can also activate the immune system, which may play a role in tumor control. Viruses can be modified in a variety of ways to improve their selectivity and efficacy. The adenovirus genome can be easily engineered to incorporate different tumor targeting mechanisms and therapeutic transgenes for improved antitumor properties. Here we review the available preclinical and clinical data on use of oncolytic adenoviruses in humans.


Asunto(s)
Adenoviridae/fisiología , Neoplasias/terapia , Virus Oncolíticos/fisiología , Adenoviridae/genética , Terapia Genética , Humanos , Viroterapia Oncolítica , Virus Oncolíticos/genética
13.
Mol Pharm ; 8(1): 93-103, 2011 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-20964369

RESUMEN

Oncolytic adenoviruses are an emerging treatment option for advanced and refractory cancer. Such patients are often treated with corticosteroids to ameliorate tumor associated symptoms. Thus, it is important to evaluate whether safety is affected by immunosuppression possibly induced by corticosteroids. Concurrent low-dose cyclophosphamide, appealing for its immunomodulatory effects, could also impact safety. In a retrospective case-control study, we evaluated the effect of systemic corticosteroid use in cancer patients receiving oncolytic virotherapy. Four treatment groups were identified: (1) oncolytic adenovirus with oral glucocorticoids, (2) virus alone, (3) virus with glucocorticoids and cyclophosphamide and (4) virus with cyclophosphamide. Adverse events, neutralizing antibody titers, viral DNA in circulation and tumor responses were evaluated. The most common adverse effects were grade 1-2 fatigue, nausea, fever and abdominal pain. Common asymptomatic findings included self-limiting grade 1-3 hyponatremia and aspartate aminotransferase increase. Safety was good and no significant differences were observed between the groups. All patients had an increase in neutralizing antibody titers post-treatment, and no trends for differences between groups were observed. There were fewer post-treatment virus genomes circulating in patients receiving glucocorticoids when compared to their control groups. Overall, glucocorticoid use in cancer patients receiving oncolytic adenovirus, with or without low-dose cyclophosphamide, seems safe.


Asunto(s)
Adenoviridae/genética , Glucocorticoides/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Viroterapia Oncolítica/efectos adversos , Estudios Retrospectivos , Resultado del Tratamiento
14.
Int J Radiat Oncol Biol Phys ; 78(4): 1201-9, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20832189

RESUMEN

PURPOSE: Radiotherapy is widely used for treatment of many tumor types, but it can damage normal tissues. It has been proposed that cancer cells can be selectively sensitized to radiation by adenovirus replication or by using radiosensitizing transgenes. Adenoviral proteins E1B55K, E4orf3, and E4orf6 play a role in radiosensitization, by targeting the Mre11, Rad50, and NBS1 complex (MRN) and inhibiting DNA double-strand break (DSB) repair. We hypothesize that combined with irradiation, these adenoviral proteins increase cell killing through the impairment of DSB repair. METHODS AND MATERIALS: We assessed the radiosensitizing/additive potential of replication-deficient adenoviruses expressing E1B55K, E4orf3, and E4orf6 proteins. Combination treatments with low-dose external photon beam radiotherapy were studied in prostate cancer (PC-3MM2 and DU-145), breast cancer (M4A4-LM3), and head and neck cancer (UT-SCC8) cell lines. We further demonstrated radiosensitizing or additive effects in mice with PC-3MM2 tumors. RESULTS: We show enhanced cell killing with adenovirus and radiation combination treatment. Co-infection with several of the viruses did not further increase cell killing, suggesting that both E4orf6 and E4orf3 are potent in MRN inhibition. Our results show that adenoviral proteins E4orf3 and E4orf6, but not E1B55K, are effective also in vivo. Enhanced cell killing was due to inhibition of DSB repair resulting in persistent double-strand DNA damage, indicated by elevated phospho-H2AX levels at 24 h after irradiation. CONCLUSIONS: This knowledge can be applied for improving the treatment of malignant tumors, such as prostate cancer, for development of more effective combination therapies and minimizing radiation doses and reducing side effects.


Asunto(s)
Proteínas E4 de Adenovirus/fisiología , Adenovirus Humanos/metabolismo , Neoplasias/radioterapia , Tolerancia a Radiación/fisiología , Proteínas Virales/fisiología , Proteínas E4 de Adenovirus/metabolismo , Adenovirus Humanos/genética , Animales , Neoplasias de la Mama/radioterapia , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Reparación del ADN , Femenino , Neoplasias de Cabeza y Cuello/radioterapia , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/virología , Neoplasias de la Próstata/radioterapia , Dosificación Radioterapéutica , Distribución Aleatoria , Proteínas Virales/metabolismo
15.
Mol Ther ; 18(10): 1874-84, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20664527

RESUMEN

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.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adolescente , Adulto , Anciano , Animales , Línea Celular , Línea Celular Tumoral , Cricetinae , Ciclofosfamida/uso terapéutico , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Inmunosupresores/uso terapéutico , Masculino , Mesocricetus , Persona de Mediana Edad , Neoplasias/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
16.
Cancer Res ; 70(11): 4297-309, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20484030

RESUMEN

Granulocyte macrophage colony-stimulating factor (GMCSF) can mediate antitumor effects by recruiting natural killer cells and by induction of tumor-specific cytotoxic T-cells through antigen-presenting cells. Oncolytic tumor cell-killing can produce a potent costimulatory danger signal and release of tumor epitopes for antigen-presenting cell sampling. Therefore, an oncolytic adenovirus coding for GMCSF was engineered and shown to induce tumor-specific immunity in an immunocompetent syngeneic hamster model. Subsequently, 20 patients with advanced solid tumors refractory to standard therapies were treated with Ad5-D24-GMCSF. Of the 16 radiologically evaluable patients, 2 had complete responses, 1 had a minor response, and 5 had disease stabilization. Responses were frequently seen in injected and noninjected tumors. Treatment was well tolerated and resulted in the induction of both tumor-specific and virus-specific immunity as measured by ELISPOT and pentamer analysis. This is the first time that oncolytic virus-mediated antitumor immunity has been shown in humans. Ad5-D24-GMCSF is promising for further clinical testing.


Asunto(s)
Adenoviridae/genética , Factor Estimulante de Colonias de Granulocitos/genética , Inmunoterapia/métodos , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adenoviridae/inmunología , Adenoviridae/metabolismo , Animales , Cricetinae , Epítopos de Linfocito T/inmunología , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Factor Estimulante de Colonias de Granulocitos/inmunología , Humanos , Proteínas Inhibidoras de la Apoptosis , Proteínas Asociadas a Microtúbulos/inmunología , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/virología , Survivin , Linfocitos T/inmunología , Transfección
17.
Clin Cancer Res ; 16(11): 3035-43, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20501623

RESUMEN

PURPOSE: Twenty-one patients with cancer were treated with a single round of oncolytic adenovirus ICOVIR-7. EXPERIMENTAL DESIGN: ICOVIR-7 features an RGD-4C modification of the fiber HI-loop of serotype 5 adenovirus for enhanced entry into tumor cells. Tumor selectivity is mediated by an insulator, a modified E2F promoter, and a Rb-binding site deletion of E1A, whereas replication is optimized with E2F binding hairpins and a Kozak sequence. ICOVIR-7 doses ranged from 2 x 10(10) to 1 x 10(12) viral particles. All patients had advanced and metastatic solid tumors refractory to standard therapies. RESULTS: ICOVIR-7 treatment was well tolerated with mild to moderate fever, fatigue, elevated liver transaminases, chills, and hyponatremia. One patient had grade 3 anemia but no other serious side effects were seen. At baseline, 9 of 21 of patients had neutralizing antibody titers against the ICOVIR-7 capsid. Treatment resulted in neutralizing antibody titer induction within 4 weeks in 16 of 18 patients. No elevations of serum proinflammatory cytokine levels were detected. Viral genomes were detected in the circulation in 18 of 21 of patients after injection and 7 of 15 of the samples were positive 2 to 4 weeks later suggesting viral replication. CONCLUSIONS: Overall, objective evidence of antitumor activity was seen in 9 of 17 evaluable patients. In radiological analyses, 5 of 12 evaluable patients had stabilization or reduction in tumor size. These consisted of one partial response, two minor responses and two cases of stable disease, all occurring in patients who had progressive disease before treatment. In summary, ICOVIR-7 treatment is apparently safe, resulting in anticancer activity, and is therefore promising for further clinical testing.


Asunto(s)
Adenoviridae , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adolescente , Adulto , Anciano , Anticuerpos Antivirales/análisis , Niño , Femenino , Humanos , Interleucinas/sangre , Masculino , Persona de Mediana Edad , Viroterapia Oncolítica/efectos adversos , Retratamiento , Resultado del Tratamiento , Replicación Viral
18.
PLoS One ; 3(8): e2917, 2008 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-18698374

RESUMEN

Clinical trials have confirmed the safety of selectively oncolytic adenoviruses for treatment of advanced cancers. However, increasingly effective viruses could result in more toxicity and therefore it would be useful if replication could be abrogated if necessary. We analyzed viruses containing the cyclooxygenase-2 (Cox-2) or vascular endothelial growth factor (VEGF) promoter for controlling replication. Anti-inflammatory agents can lower Cox-2 protein levels and therefore we hypothesized that also the promoter might be affected. As Cox-2 modulates expression of VEGF, also the VEGF promoter might be controllable. First, we evaluated the effect of anti-inflammatory agents on promoter activity or adenovirus infectivity in vitro. Further, we analyzed the oncolytic potency of the viruses in vitro and in vivo with and without the reagents. Moreover, the effect of on virus replication was analyzed. We found that RGD-4C or Ad5/3 modified fibers improved the oncolytic potency of the viruses in vitro and in vivo. We found that both promoters could be downregulated with dexamethasone, sodium salicylate, or salicylic acid. Oncolytic efficacy correlated with the promoter activity and in vitro virus production could be abrogated with the substances. In vivo, we saw good therapeutic efficacy of the viruses in a model of intravenous therapy of metastatic cervical cancer, but the inhibitory effect of dexamethasone was not strong enough to provide significant differences in a complex in vivo environment. Our results suggest that anti-inflammatory drugs may affect the replication of adenovirus, which might be relevant in case of replication associated side effects.


Asunto(s)
Adenoviridae , Viroterapia Oncolítica/métodos , Neoplasias del Cuello Uterino/rehabilitación , Neoplasias del Cuello Uterino/virología , División Celular , Línea Celular Tumoral , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Femenino , Células HeLa , Humanos , Virus Oncolíticos/genética , Regiones Promotoras Genéticas , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Factor A de Crecimiento Endotelial Vascular/genética , Replicación Viral/genética
19.
Cancer Res ; 68(14): 5533-9, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18632604

RESUMEN

It has been proposed that human tumors contain stem cells that have a central role in tumor initiation and posttreatment relapse. Putative breast cancer stem cells may reside in the CD44(+)CD24(-/low) population. Oncolytic adenoviruses are attractive for killing of these cells because they enter through infection and are therefore not susceptible to active and passive mechanisms that render stem cells resistant to many drugs. Although adenoviruses have been quite safe in cancer trials, preclinical work suggests that toxicity may eventually be possible with more active agents. Therefore, restriction of virus replication to target tissues with tissues-specific promoters is appealing for improving safety and can be achieved without loss of efficacy. We extracted CD44(+)CD24(-/low) cells from pleural effusions of breast cancer patients and found that modification of adenovirus type 5 tropism with the serotype 3 knob increased gene delivery to CD44(+)CD24(-/low) cells. alpha-Lactalbumin, cyclo-oxygenase 2, telomerase, and multidrug resistance protein promoters were studied for activity in CD44(+)CD24(-/low) cells, and a panel of oncolytic viruses was subsequently constructed. Each virus featured 5/3 chimerism of the fiber and a promoter controlling expression of E1A, which was also deleted in the Rb binding domain for additional tumor selectivity. Cell killing assays identified Ad5/3-cox2L-d24 and Ad5/3-mdr-d24 as the most active agents, and these viruses were able to completely eradicate CD44(+)CD24(-/low) cells in vitro. In vivo, these viruses had significant antitumor activity in CD44(+)CD24(-/low)-derived tumors. These findings may have relevance for elimination of cancer stem cells in humans.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Antígeno CD24/biosíntesis , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Receptores de Hialuranos/biosíntesis , Regiones Promotoras Genéticas , Proteínas E1A de Adenovirus/metabolismo , Animales , Antineoplásicos/uso terapéutico , Femenino , Citometría de Flujo/métodos , Humanos , Ratones , Viroterapia Oncolítica/métodos , Virus Oncolíticos/metabolismo
20.
Mol Cancer Ther ; 6(2): 742-51, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17308070

RESUMEN

Hormone refractory metastatic prostate cancer is a deadly disease that currently lacks curative treatments. Conditionally replicating adenoviruses (CRAds) are promising new agents against cancer due to their innate capability to cause oncolysis of tumor cells. Their antitumor effect is determined in part by their capacity for infecting cancer cells. However, the respective primary receptor, the coxsackie-adenovirus receptor (CAR), is variably expressed in many cancer types. We created Ad5/3Delta24hCG, a novel CRAd retargeted to the adenovirus serotype 3 receptor, which has been reported to be highly expressed in tumors. Furthermore, we added a transgene for the beta-chain of human chorionic gonadotropin (hCGbeta), whose expression was tightly coupled to virus replication. Ad5/3Delta24hCG was found effective in killing prostate cancer cells, and oncolysis was seen in concordance with hCGbeta production. In a s.c. in vivo model of hormone refractory prostate cancer, Ad5/3Delta24hCG treatment resulted in statistically significant tumor growth inhibition. Moreover, i.v. injection of Ad5/3Delta24hCG prolonged the survival of mice with hormone refractory prostate cancer metastatic to the lung. Detection of hCGbeta in serum samples confirmed viral replication in vivo. Infection of human clinical samples of cancerous and normal prostatic tissue resulted in effective hCGbeta production in cancer tissue, whereas it remained low in nonmalignant tissue, suggesting cancer-specific replication. These results suggest that Ad5/3Delta24hCG is a potent virus for the treatment of hormone refractory prostate cancer in vitro and in vivo. These preclinical data set the stage for translation into clinical studies.


Asunto(s)
Adenoviridae/fisiología , Gonadotropina Coriónica/farmacología , Neoplasias Pulmonares/terapia , Viroterapia Oncolítica , Neoplasias de la Próstata/terapia , Replicación Viral , Animales , Supervivencia Celular , Células Cultivadas , Gonadotropina Coriónica/genética , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , ADN Viral/genética , Terapia Genética , Vectores Genéticos , Humanos , Riñón/metabolismo , Riñón/virología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/virología , Masculino , Ratones , Ratones Desnudos , Oligopéptidos/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/virología , Receptores Virales/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...