Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cancer Res ; 82(5): 916-928, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34965932

RESUMO

Squamous cell carcinoma driven by human papillomavirus (HPV) is more sensitive to DNA-damaging therapies than its HPV-negative counterpart. Here, we show that p16, the clinically used surrogate for HPV positivity, renders cells more sensitive to radiotherapy via a ubiquitin-dependent signaling pathway, linking high levels of this protein to increased activity of the transcription factor SP1, increased HUWE1 transcription, and degradation of ubiquitin-specific protease 7 (USP7) and TRIP12. Activation of this pathway in HPV-positive disease led to decreased homologous recombination and improved response to radiotherapy, a phenomenon that can be recapitulated in HPV-negative disease using USP7 inhibitors in clinical development. This p16-driven axis induced sensitivity to PARP inhibition and potentially leads to "BRCAness" in head and neck squamous cell carcinoma (HNSCC) cells. Thus, these findings support a functional role for p16 in HPV-positive tumors in driving response to DNA damage, which can be exploited to improve outcomes in both patients with HPV-positive and HPV-negative HNSCC. SIGNIFICANCE: In HPV-positive tumors, a previously undiscovered pathway directly links p16 to DNA damage repair and sensitivity to radiotherapy via a clinically relevant and pharmacologically targetable ubiquitin-mediated degradation pathway.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Infecções por Papillomavirus , Carcinoma de Células Escamosas/patologia , Proteínas de Transporte , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Dano ao DNA , DNA Viral/genética , Neoplasias de Cabeça e Pescoço/genética , Humanos , Papillomaviridae/genética , Transdução de Sinais , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina , Ubiquitina-Proteína Ligases/metabolismo , Peptidase 7 Específica de Ubiquitina/metabolismo
2.
Int J Radiat Biol ; 97(8): 1121-1128, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32073931

RESUMO

PURPOSE: Head and neck cancers (HNSCC) are routinely treated with radiotherapy; however, normal tissue toxicity remains a concern. Therefore, it is important to validate treatment modalities combining molecularly targeted agents with radiotherapy to improve the therapeutic ratio. The aim of this study was to assess the ability of the PARP inhibitor niraparib (MK-4827) alone, or in combination with cell cycle checkpoint abrogating drugs targeting Chk1 (MK-8776) or Wee1 (MK-1775), to radiosensitize HNSCCs in the context of HPV status. MATERIALS AND METHODS: PARP1, PARP2, Chk1 or Wee1 shRNA constructs were analyzed from an in vivo shRNA screen of HNSCC xenografts comparing radiosensitization differences between HPV(+) and HPV(-) tumors. Radiosensitization by niraparib alone or in combination with MK-8776 or MK-1775 was assessed by clonogenic survival in HPV(-) and HPV(+) cells; and the role of p16 in determining response was explored. Relative expressions of DNA repair genes were compared by PCR array in HPV(+) and HPV(-) cells, and following siRNA-mediated knockdown of TRIP12 in HPV(-) cells. RESULTS: In vivo shRNA screening showed a modest preferential radiosensitization by Wee1 and PARP2 in HPV(-) and Chk1 in HPV(+) tumor models. Niraparib alone enhanced the radiosensitivity of all HNSCC cell lines tested. However, HPV(-) cells were sensitized to a greater degree, as suggested by the shRNA screen. When combined with MK-8776 or MK-1775, radiosensitization was further enhanced in an HPV dependent manner with HPV(+) cells enhanced by MK-8776 and HPV(-) cells enhanced by MK-1775. A PCR array for DNA repair genes showed PARP and HR proteins BRCA1 and RAD51 were much lower in HPV(+) cells than in HPV(-). Similarly, directly knocking down p16-dependent TRIP12 decreased expression of these same genes. Overexpressing p16 decreased TRIP12 expression and increased radiosensitivity in HPV(-) HN5. However, while PARP inhibition led to significant radiosensitization in the control, it led to no further significant radiosensitization in p16 overexpressing cells. Forced p16 expression in HPV(-) HN5 increased accumulation in G1 and subG1 and limited progression to S phase, thus reducing effectiveness of PARP inhibition. CONCLUSIONS: Niraparib effectively radiosensitizes HNSCCs with a greater benefit seen in HPV(-). HPV status also plays a role in response to MK-8776 or MK-1775 when combined with niraparib due to differences in DNA repair mechanisms. This study suggests that using cell cycle abrogators in combination with PARP inhibitors may be a beneficial treatment option in HNSCC, but also emphasizes the importance of HPV status when considering effective treatment strategies.


Assuntos
Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Dano ao DNA , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Humanos , Indazóis/farmacologia , Piperidinas/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Pirazóis/farmacologia , Pirimidinonas/farmacologia , Tolerância a Radiação/efeitos dos fármacos
3.
Sci Rep ; 9(1): 1949, 2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30760738

RESUMO

Unresectable pancreatic cancer is almost universally lethal because chemotherapy and radiation cannot completely stop the growth of the cancer. The major problem with using radiation to approximate surgery in unresectable disease is that the radiation dose required to ablate pancreatic cancer exceeds the tolerance of the nearby duodenum. WR-2721, also known as amifostine, is a well-known radioprotector, but has significant clinical toxicities when given systemically. WR-2721 is a prodrug and is converted to its active metabolite, WR-1065, by alkaline phosphatases in normal tissues. The small intestine is highly enriched in these activating enzymes, and thus we reasoned that oral administration of WR-2721 just before radiation would result in localized production of the radioprotective WR-1065 in the small intestine, providing protective benefits without the significant systemic side effects. Here, we show that oral WR-2721 is as effective as intraperitoneal WR-2721 in promoting survival of intestinal crypt clonogens after morbid irradiation. Furthermore, oral WR-2721 confers full radioprotection and survival after lethal upper abdominal irradiation of 12.5 Gy × 5 fractions (total of 62.5 Gy, EQD2 = 140.6 Gy). This radioprotection enables ablative radiation therapy in a mouse model of pancreatic cancer and nearly triples the median survival compared to controls. We find that the efficacy of oral WR-2721 stems from its selective accumulation in the intestine, but not in tumors or other normal tissues, as determined by in vivo mass spectrometry analysis. Thus, we demonstrate that oral WR-2721 is a well-tolerated, and quantitatively selective, radioprotector of the intestinal tract that is capable of enabling clinically relevant ablative doses of radiation to the upper abdomen without unacceptable gastrointestinal toxicity.


Assuntos
Amifostina/farmacologia , Mercaptoetilaminas/farmacologia , Protetores contra Radiação/uso terapêutico , Administração Oral , Amifostina/metabolismo , Animais , Feminino , Intestino Delgado/efeitos dos fármacos , Masculino , Mercaptoetilaminas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/tratamento farmacológico , Doses de Radiação , Proteção Radiológica/métodos , Neoplasias Pancreáticas
4.
Clin Cancer Res ; 13(6): 1868-74, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17363543

RESUMO

PURPOSE: 130-nm albumin-bound paclitaxel (nab-paclitaxel) is a novel solvent-free albumin-bound paclitaxel, designed to avoid solvent-related toxicity. Nab-paclitaxel has been successfully introduced into the clinic but its radiation-enhancing potential has not yet been evaluated. We conducted a preclinical evaluation of the radiation-modulating effects of nab-paclitaxel in tumor and normal tissues. EXPERIMENTAL DESIGN: Mice bearing syngeneic ovarian or mammary carcinomas were treated with nab-paclitaxel, radiation, or combination of both. Nab-paclitaxel was administered at 90 mg/kg, 1.5 times the maximum tolerated dose for solvent-based paclitaxel. End points were antitumor efficacy (growth delay, radiocurability, and cellular effects) and normal tissue toxicity (gut and skin). RESULTS: Nab-paclitaxel showed single-agent antitumor efficacy against both tumor types and acted as a radiosensitizer. Combined with radiation, nab-paclitaxel produced supra-additive effects when given before radiation. Nab-paclitaxel significantly increased radiocurability by reducing the dose yielding 50% tumor cure (TCD(50)) from 54.3 to 35.2 Gy. Tumor histology following nab-paclitaxel treatment was characterized by pronounced necrotic and apoptotic cell death and mitotic arrest. Nab-paclitaxel did not increase normal tissue radioresponse. CONCLUSIONS: Nab-paclitaxel exhibited strong antitumor efficacy against both tumors as a single agent and it improved radiotherapy in a supra-additive manner. These improved effects were achieved without increased normal tissue toxicity to either rapidly or slowly proliferating normal tissues although the drug dose was 1.5 times higher than the maximum tolerated dose of solvent-based paclitaxel. These preclinical findings show that combining nab-paclitaxel with radiotherapy would improve the outcome of taxane-based chemoradiotherapy. This novel taxane is thus a good candidate for testing in clinical chemoradiotherapy trials.


Assuntos
Adenocarcinoma/radioterapia , Albuminas/uso terapêutico , Neoplasias Mamárias Experimentais/radioterapia , Neoplasias Ovarianas/radioterapia , Paclitaxel/uso terapêutico , Radiossensibilizantes/uso terapêutico , Adenocarcinoma/patologia , Albuminas/química , Animais , Antineoplásicos/uso terapêutico , Terapia Combinada , Feminino , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias , Neoplasias Ovarianas/patologia , Paclitaxel/química , Doses de Radiação , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/efeitos da radiação
5.
Int J Radiat Oncol Biol Phys ; 68(4): 968-70, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17512131

RESUMO

The clinical use of protons for the treatment of cancer is one of the most technologically advanced forms of therapy. The newly commissioned M.D. Anderson Cancer Center, Proton Therapy Center Houston is one of the newest facilities offering a complete range of advanced proton therapies designed specifically to minimize treatment-related normal tissue toxicity. As an integral part of the clinical commissioning of this proton beam, we performed an in vivo assessment of the relative biologic effectiveness of the 250-MeV protons relative to standard cobalt(60) using the jejunal crypt microcolony assay in mice. Our data are consistent with a generic relative biologic effectiveness of 1.1, and this relative biologic effectiveness is in agreement with that used to describe most other clinical proton therapy beams worldwide.


Assuntos
Hospitais Especializados , Jejuno/efeitos da radiação , Terapia com Prótons , Eficiência Biológica Relativa , Animais , Sobrevivência Celular/efeitos da radiação , Radioisótopos de Cobalto/uso terapêutico , Relação Dose-Resposta à Radiação , Feminino , Jejuno/citologia , Camundongos , Camundongos Endogâmicos C3H , Texas
6.
Drugs ; 67(6): 821-45, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17428102

RESUMO

Despite recent improvements in chemotherapy and radiation therapy in cancer management with the addition of biological agents, novel treatment approaches are needed to further benefit patients. Cyclo-oxygenase (COX)-2 inhibition represents one such possibility. COX-2 is an enzyme induced in pathological states such as inflammatory disorders and cancer, where it mediates production of prostanoids. The enzyme is commonly expressed in both premalignant lesions and malignant tumours of different types. A growing body of evidence suggests an association of COX-2 with tumour development, aggressive biological tumour behaviour, resistance to standard cancer treatment, and adverse patient outcome. COX-2 may be related to cancer development and propagation through multiple mechanisms, including stimulation of growth, migration, invasiveness, resistance to apoptosis, suppression of the immunosurveillance system, and enhancement of angiogenesis. Epidemiological data suggest that NSAIDs and selective COX-2 inhibitors might prevent the development of cancers, including colorectal, oesophageal and lung cancer. Preclinical investigations have demonstrated that inhibition of this enzyme with selective COX-2 inhibitors enhances tumour response to radiation and chemotherapeutic agents. These preclinical findings have been rapidly advanced to clinical oncology. Clinical trials of the combination of selective COX-2 inhibitors with radiotherapy, chemotherapy or both in patients with a number of cancers have been initiated, and preliminary results are encouraging. This review discusses the role of COX-2, its products (prostaglandins) and its inhibitors in tumour growth and treatment.


Assuntos
Antineoplásicos/farmacologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Ciclo-Oxigenase 2/fisiologia , Neoplasias/tratamento farmacológico , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Terapia Combinada , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Sistemas de Liberação de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Humanos , Neoplasias/fisiopatologia , Prostaglandinas/biossíntese , Prostaglandinas/fisiologia
7.
Radiother Oncol ; 80(2): 185-91, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16905211

RESUMO

BACKGROUND AND PURPOSE: Concurrent chemo-radiotherapy before surgery is standard treatment protocol for esophageal cancer with a less than 30% complete response due to resistance to therapy. The aim of this study was to determine whether molecular targeting approach using an inhibitor of cyclin-dependent kinases, flavopiridol, can help overcome the resistance to radiotherapy. MATERIALS AND METHODS: SEG-1 cells (human esophageal adenocarcinoma) were exposed to gamma-rays with and without flavopiridol treatment and assayed for clonogenic survival, apoptosis, cell cycle distribution, and Western blot analysis. Efficacy of flavopiridol in enhancing tumor response to radiation was determined by tumor growth delay assay using SEG-1 tumor xenografts generated in nude mice. RESULTS: The clonogenic cell survival assay data showed that flavopiridol (300 nM, 24h), when given either before or after radiation, significantly enhanced the radiosensitivity of SEG-1 cells. The cells were accumulated at G1 phase of the cell cycle by flavopiridol that was associated with downregulation of p-cdk-1, p-cdk-2, cyclin D1 and p-Rb expression. Flavopiridol by itself induced apoptosis in SEG-1 cells and also enhanced the radiation-induced apoptosis, associated with an increase in cleaved poly ADP-ribose polymerase. Reduction in phosphorylation of RNA polymerase II by flavopiridol suggested that flavopiridol inhibited the transcriptional activity. In vivo studies with SEG-1 tumor xenografts showed that flavopiridol, either given before or after radiation, greatly enhanced the effect of tumor irradiation. CONCLUSIONS: Flavopiridol treatment significantly enhanced SEG-1 cell radiosensitivity as well as the radioresponse of SEG-1 tumor xenografts. The underlying mechanisms are multiple, including cell cycle redistribution, apoptosis, and transcriptional inhibition. These preclinical data suggest that flavopiridol has the potential to increase the radioresponse of esophageal adenocarcinomas.


Assuntos
Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/radioterapia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/radioterapia , Flavonoides/farmacologia , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Adenocarcinoma/enzimologia , Adenocarcinoma/patologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Terapia Combinada , Relação Dose-Resposta a Droga , Neoplasias Esofágicas/enzimologia , Neoplasias Esofágicas/patologia , Humanos , Camundongos , Camundongos Nus , Tolerância a Radiação/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Radiother Oncol ; 80(2): 192-8, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16905212

RESUMO

BACKGROUND AND PURPOSE: Synthetic oligodeoxynucleotides (ODNs) containing unmethylated cytosine-guanine (CpG) motifs bind to Toll-like receptor 9 (TLR9) and stimulate both innate and adaptive immune reactions and possess anti-tumor activity. We recently reported that CpG ODN 1826 strongly enhances radioresponse of both immunogenic [Milas L, Mason K, Ariga H, et al. CpG oligodeoxynucleotide enhances tumor response to radiation. Cancer Res 2004;64:5074-7] and non-immunogenic [Mason KA, Ariga H, Neal R, et al. Targeting toll-like receptor-9 with CpG oligodeoxynucleotides enhances tumor response to fractionated radiotherapy. Clin Cancer Res 2005;11:361-9] murine tumors. Using two immunogenic murine tumors, a fibrosarcoma (FSa) and a mammary carcinoma (MCa-K), the present study explored whether CpG ODN 1826 also improves the response of murine tumors to the chemotherapeutic agent docetaxel (DOC). MATERIALS AND METHODS: CpG ODN 1826 (100 microg) was given sc three times: when leg tumors were 6mm, when they grew to 8mm and again 1 week later. DOC (33 mg/kg iv) and local tumor radiation (10Gy) were given when tumors were 8mm. Effects of the treatments were assayed by tumor growth delay, defined as days for tumors to grow from 8 to 12 mm in diameter. RESULTS: Treatment with CpG ODN 1826 resulted in strongly enhanced response of FSa tumors to radiation and MCa-K tumors to the chemotherapeutic agent DOC. Enhancement of tumor treatment response was demonstrated by a strong prolongation in the primary tumor treatment endpoint, tumor growth delay. Coincidentally, this treatment also resulted in a higher rate of tumor cure than that observed after tumor radiotherapy or chemotherapy alone. When all three agents were combined the effect was comparable to that of the combination of CpG ODN 1826 with radiation in the case of FSa or of the combination of CpG ODN 1826 with DOC in the case of MCa-K. CONCLUSION: Overall results show that CpG ODN 1826 can markedly improve tumor response to radiation and chemotherapy (DOC), suggesting that CpG ODNs have potential to be beneficial when used singly or in combination with other standard treatment modalities such as taxane chemotherapy, radiotherapy or both.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , DNA/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/radioterapia , Taxoides/farmacologia , Animais , Terapia Combinada , DNA/administração & dosagem , Docetaxel , Sinergismo Farmacológico , Feminino , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/efeitos da radiação , Masculino , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/radioterapia , Camundongos , Neoplasias Experimentais/genética , Oligodesoxirribonucleotídeos , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/genética , Sarcoma Experimental/patologia , Sarcoma Experimental/radioterapia , Taxoides/administração & dosagem
9.
Clin Cancer Res ; 11(1): 361-9, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15671567

RESUMO

Synthetic oligodeoxynucleotides containing unmethylated CpG motifs detected by Toll-like receptor 9 of dendritic cells and B cells have potent immunomodulatory effects. CpG oligodeoxynucleotides induce cytokines, activate natural killer cells, and elicit T-cell responses leading to antitumor effects, including improved efficacy of chemotherapeutic agents and, as we reported recently, synergy between CpG oligodeoxynucleotide 1826 and single-dose radiotherapy of an immunogenic mouse fibrosarcoma. The present study extends this finding to the fractionated radiotherapy of the fibrosarcoma tumor and assesses the ability of CpG oligodeoxynucleotide 1826 to increase the radioresponse of a tumor (nonimmunogenic fibrosarcoma). The experiments used a murine immunogenic fibrosarcoma tumor, fibrosarcoma growing in the leg of mice, and response to radiotherapy was assessed by tumor growth delay and tumor cure rate (TCD50, radiation dose yielding 50% tumor cure). Multiple s.c. peritumoral or i.t. administrations of CpG oligodeoxynucleotide 1826 at a dose of 100 microg per mouse were given when established tumors were 6 mm in diameter. Local tumor irradiation was initiated when tumors grew to 8 mm in diameter; radiation was delivered in 1 to 9 Gy fractions given twice daily separated by 6 to 7 hours for 5 consecutive days to achieve a total dose of 10 to 90 Gy. CpG oligodeoxynucleotide 1826, given as a single agent, had only a small antitumor effect, but it dramatically enhanced fibrosarcoma response to radiotherapy. Although 83.1 (79.2-90.0) Gy total dose were needed to achieve tumor cures in 50% of mice treated with radiotherapy alone, only 23.0 (11.5-32.7) Gy total dose were needed in mice treated with both CpG oligodeoxynucleotide 1826 and radiotherapy. The magnitude of potentiation of tumor radioresponse at the TCD50 level was by a factor of 3.61, a much higher value than that (a factor of 1.93) that we reported for single-dose radiotherapy. Mice cured of their tumors by combined CpG oligodeoxynucleotide 1826 plus radiotherapy were highly resistant to s.c. tumor take or development of tumor nodules in the lung from i.v. injected tumor cells when rechallenged with fibrosarcoma cells 100 to 120 days after the treatment, suggesting the development of a memory response. CpG oligodeoxynucleotide 1826 also increased radioresponse of the nonimmunogenic fibrosarcoma tumor by a factor of 1.41 and 1.73 when CpG oligodeoxynucleotide 1826 was given s.c. and i.t., respectively. These findings show that CpG oligodeoxynucleotides are highly potent enhancers of tumor response to both single-dose and fractionated radiation and as such have potential to improve clinical radiotherapy.


Assuntos
Ilhas de CpG , Proteínas de Ligação a DNA/genética , Fracionamento da Dose de Radiação , Imunoterapia/métodos , Neoplasias/genética , Neoplasias/radioterapia , Receptores de Superfície Celular/genética , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Terapia Combinada , Relação Dose-Resposta à Radiação , Fibrossarcoma/química , Fibrossarcoma/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias , Oligonucleotídeos/química , Fatores de Tempo , Receptor Toll-Like 9
10.
Cancer Res ; 64(15): 5074-7, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15289307

RESUMO

CpG oligodeoxynucleotides (ODNs) are synthetic DNA sequences containing unmethylated cytosine-guanine motifs with potent immunomodulatory effects. Via Toll-like receptor 9 agonism of dendritic cells and B cells, CpG ODNs induce cytokines, activate natural killer cells, and elicit vigorous T-cell responses that lead to significant antitumor effects, including improved efficacy of chemotherapeutic agents. On the basis of these properties of CpG ODNs, we tested whether they also could enhance tumor response to radiotherapy. Using an immunogenic mouse tumor, designated FSa, the response to radiotherapy was assayed by tumor growth delay and tumor cure rate (TCD(50), radiation dose yielding 50% tumor cure rate). Treatments were initiated when established tumors were either 6 or 8 mm in diameter. CpG ODN as a single agent given s.c. peritumorally had little effect on tumor growth; however, it dramatically enhanced tumor growth delay in response to single-dose radiation by a factor of 2.58-2.65. CpG ODN also dramatically improved tumor radiocurability, reducing the TCD(50) by a factor of 1.93, from 39.6 (36.1-43.1) Gy to 20.5 (14.3-25.7) Gy. The CpG ODN-induced enhancement of tumor radioresponse was diminished in tumor-bearing mice immunocompromised by sublethal whole-body radiation. Tumors treated with CpG ODN and radiation showed histologic changes characterized by increased necrosis, heavy infiltration by host inflammatory cells (lymphocytes and granulocytes), and reduced tumor cell density. These results show that CpG ODNs are potent enhancers of tumor radioresponse and as such have potential to improve clinical radiotherapy.


Assuntos
Oligodesoxirribonucleotídeos/farmacologia , Radiossensibilizantes/uso terapêutico , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/radioterapia , Animais , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Metilcolantreno , Camundongos , Camundongos Endogâmicos C3H , Necrose , Doses de Radiação , Sarcoma Experimental/imunologia , Linfócitos T/imunologia , Irradiação Corporal Total
11.
Int J Radiat Biol ; 92(12): 855-857, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27807995

RESUMO

We have collected lab notebooks from Rod Wither's many years of experimentation, from laboratories in Houston and Los Angeles, as well as from several of his collaborators in the USA and overseas. The contents have been digitized, and in this note we explain the mechanism that has been set up to make the 'Withers Archive' available online.


Assuntos
Arquivos/história , Manuscritos Médicos como Assunto/história , Radioterapia (Especialidade)/história , Radiobiologia/história , Austrália , História do Século XX , História do Século XXI , Sistemas On-Line , Estados Unidos
12.
Int J Radiat Oncol Biol Phys ; 92(4): 829-36, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25956832

RESUMO

PURPOSE: To investigate differences in tumor histotype, incidence, latency, and strain susceptibility in mice exposed to single-dose or clinically relevant, fractioned-dose γ-ray radiation. METHODS AND MATERIALS: C3Hf/Kam and C57BL/6J mice were locally irradiated to the right hindlimb with either single large doses between 10 and 70 Gy or fractionated doses totaling 40 to 80 Gy delivered at 2-Gy/d fractions, 5 d/wk, for 4 to 8 weeks. The mice were closely evaluated for tumor development in the irradiated field for 800 days after irradiation, and all tumors were characterized histologically. RESULTS: A total of 210 tumors were induced within the radiation field in 788 mice. An overall decrease in tumor incidence was observed after fractionated irradiation (16.4%) in comparison with single-dose irradiation (36.1%). Sarcomas were the predominant postirradiation tumor observed (n=201), with carcinomas occurring less frequently (n=9). The proportion of mice developing tumors increased significantly with total dose for both single-dose and fractionated schedules, and latencies were significantly decreased in mice exposed to larger total doses. C3Hf/Kam mice were more susceptible to tumor induction than C57BL/6J mice after single-dose irradiation; however, significant differences in tumor susceptibilities after fractionated radiation were not observed. For both strains of mice, osteosarcomas and hemangiosarcomas were significantly more common after fractionated irradiation, whereas fibrosarcomas and malignant fibrous histiocytomas were significantly more common after single-dose irradiation. CONCLUSIONS: This study investigated the tumorigenic effect of acute large doses in comparison with fractionated radiation in which both the dose and delivery schedule were similar to those used in clinical radiation therapy. Differences in tumor histotype after single-dose or fractionated radiation exposures provide novel in vivo evidence for differences in tumor susceptibility among stromal cell populations.


Assuntos
Carcinoma/patologia , Neoplasias Induzidas por Radiação/patologia , Sarcoma/patologia , Animais , Carcinoma/etiologia , Carcinoma de Células Escamosas/etiologia , Carcinoma de Células Escamosas/patologia , Fracionamento da Dose de Radiação , Fibrossarcoma/etiologia , Fibrossarcoma/patologia , Hemangiossarcoma/etiologia , Hemangiossarcoma/patologia , Histiocitoma Fibroso Maligno/etiologia , Histiocitoma Fibroso Maligno/patologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Doses de Radiação , Sarcoma/etiologia
13.
Cancer Med ; 4(1): 65-74, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25355701

RESUMO

Interaction between the epidermal growth factor receptor (EGFR) and the insulin-like growth factor receptor (IGF-1R) has been well established in many cancer types. We investigated the effects of cetuximab (EGFR antibody) and IMC-A12 (IGF-1R antibody) on the response of head and neck squamous cell carcinoma (HNSCC) to radiation therapy (RT). The effects of cetuximab and IMC-A12 on cell viability and radiosensitivity were determined by clonogenic cell survival assay. Formation of nuclear γ-H2AX and 53BP1 foci was monitored by immunofluorescence. Alterations in target signaling were analyzed by Western blots. In vivo tumor growth delay assay was performed to determine the efficacy of triple therapy with IMC-A12, cetuximab, and RT. In vitro data showed that cetuximab differentially affected the survival and the radiosensitivity of HNSCC cells. Cetuximab suppressed DNA repair that was evident by the prolonged presence of nuclear γ-H2AX and 53BP1 foci. IMC-A12 did not have any effect on the cell survival. However, it increased the radiosensitivity of one of the cell lines. EGFR inhibition increased IGF-1R expression levels and also the association between EGFR and IGF-1R. Addition of IMC-A12 to cetuximab did not increase the radiosensitivity of these cells. Tumor xenografts exhibited enhanced response to RT in the presence of either cetuximab or IMC-A12. Concurrent treatment regimen failed to further enhance the tumor response to cetuximab and/or RT. Taken together our data suggest that concomitant inhibition of both EGFR and IGF-1R pathways did not yield additional therapeutic benefit in overcoming resistance to RT.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Receptores ErbB/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cetuximab , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Modelos Animais de Doenças , Receptores ErbB/genética , Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/mortalidade , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/terapia , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Multimerização Proteica/efeitos dos fármacos , Radiação , Tolerância a Radiação/efeitos dos fármacos , Radioterapia , Receptor IGF Tipo 1/genética , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/efeitos da radiação , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Int J Radiat Oncol Biol Phys ; 55(3): 707-12, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12573758

RESUMO

PURPOSE: Conjugating drugs with polymeric carriers is one way to improve selective delivery to tumors. Poly (L-glutamic acid)-paclitaxel (PG-TXL) is one such conjugate. Compared with paclitaxel, its uptake, tumor retention, and antitumor efficacy are increased. Initial studies showed that PG-TXL given 24 h before or after radiotherapy enhanced tumor growth delay significantly more than paclitaxel. To determine if PG-TXL-induced enhancement is obtained in a more clinically relevant setting, we investigated PG-TXL effects on tumor cure. METHODS AND MATERIALS: Mice bearing 7-mm-diameter ovarian carcinomas were treated with PG-TXL at an equivalent paclitaxel dose of 80 mg/kg, single dose or 5 daily fractions of radiation or both PG-TXL and radiation. Treatment endpoint was TCD(50) (radiation dose yielding tumor control in 50% of mice). Acute radioresponse of jejunum, skin, and hair was determined for all treatments. RESULTS: PG-TXL dramatically improved tumor radioresponse, reducing TCD(50) of single-dose irradiation from 53.9 (52.2-55.5) Gy to 7.5 (4.5-10.7) Gy, an enhancement factor (EF) of 7.2. The drug improved the efficacy of fractionated irradiation even more, reducing the TCD(50) of 66.6 (62.8-90.4) Gy total fractionated dose to only 7.9 (4.3-11.5) Gy, for an EF of 8.4. PG-TXL did not affect normal tissue radioresponse resulting from either single or fractionated irradiation. CONCLUSION: PG-TXL dramatically potentiated tumor radiocurability after single-dose or fractionated irradiation without affecting acute normal tissue injury. To our knowledge, PG-TXL increased the therapeutic ratio of radiotherapy more than that previously reported for other taxanes, thus, PG-TXL has a high potential to improve clinical radiotherapy.


Assuntos
Adenocarcinoma/radioterapia , Neoplasias Ovarianas/radioterapia , Paclitaxel/análogos & derivados , Paclitaxel/uso terapêutico , Ácido Poliglutâmico/uso terapêutico , Radiossensibilizantes/uso terapêutico , Taxoides , Adenocarcinoma/tratamento farmacológico , Animais , Terapia Combinada , Intervalos de Confiança , Fracionamento da Dose de Radiação , Relação Dose-Resposta à Radiação , Feminino , Cabelo/efeitos da radiação , Infusões Intravenosas , Doenças do Jejuno/etiologia , Camundongos , Camundongos Endogâmicos C3H , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/administração & dosagem , Ácido Poliglutâmico/administração & dosagem , Lesões por Radiação/etiologia , Radiossensibilizantes/administração & dosagem , Radiodermite/etiologia , Dosagem Radioterapêutica , Organismos Livres de Patógenos Específicos
15.
Int J Radiat Oncol Biol Phys ; 58(2): 369-75, 2004 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-14751505

RESUMO

Cyclooxygenase-2 (COX-2) is an enzyme expressed primarily in pathologic states, such as inflammatory disorders and cancer, where it mediates prostaglandin production. Its overexpression is associated with more aggressive biologic tumor behavior and adverse patient outcome. Increasing evidence shows that agents that selectively inhibit COX-2 enhance tumor response to radiation or chemotherapeutic agents. This article gives an overview of some of this evidence. In addition, we describe new results showing that celecoxib, a selective COX-2 inhibitor, enhanced response of A431 human tumor xenografts in nude mice to radiation by an enhancement factor (EF) of 1.43 and to the chemotherapeutic agent docetaxel by an EF of 2.07. Celecoxib also enhanced tumor response when added to the combined docetaxel plus radiation treatment (EF = 2.13). Further experiments showed that selective COX-2 inhibitors enhanced tumor cell sensitivity to ionizing radiation, involving inhibition of cellular repair from radiation damage and cell cycle redistribution as mechanisms for some cell types. The results show that selective COX-2 inhibitors have the potential to improve tumor radiotherapy or radiochemotherapy, and this therapeutic strategy is currently under clinical testing.


Assuntos
Inibidores de Ciclo-Oxigenase/uso terapêutico , Isoenzimas/antagonistas & inibidores , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Pirazóis/uso terapêutico , Radiossensibilizantes/uso terapêutico , Sulfonamidas/uso terapêutico , Animais , Celecoxib , Terapia Combinada , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Interações Medicamentosas , Humanos , Isoenzimas/metabolismo , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C3H , Camundongos Nus , Proteínas de Neoplasias/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Tolerância a Radiação
16.
Clin Lung Cancer ; 4(6): 356-65, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-14599301

RESUMO

Cyclooxygenase-2 (COX-2) is an enzyme involved in prostaglandin production in pathologic states such as inflammatory disorders and cancer. The enzyme is often overexpressed in premalignant lesions and cancer of the lung. Overexpression of COX-2 in lung cancer is associated with more aggressive biological tumor behavior and adverse patient outcome. In preclinical studies, inhibition of this enzyme with selective COX-2 inhibitors enhances tumor response to radiation and chemotherapeutic agents. These findings have been rapidly advanced to clinical oncology. Clinical trials of the combination of selective COX-2 inhibitors with radiation therapy, chemotherapy, or both in patients with lung cancer have been initiated and some preliminary results are available. In this review, we describe the relationship between overexpression of COX-2 and lung cancer, the antitumor effect of selective COX-2 inhibitors, discuss the rationale for using selective COX-2 inhibitors combined with radiation therapy and chemotherapy, and summarize current clinical protocols and initial findings.

17.
Clin Lung Cancer ; 5(6): 340-52, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15217533

RESUMO

Despite considerable efforts to reduce tobacco use, lung cancer remains the most common cancer in both men and women. Recent advances in radiation therapy and chemotherapy for lung cancer have yielded encouraging results, but survival in patients with locally advanced non-small-cell lung cancer (NSCLC) remains poor. As more and more molecular changes and their importance in malignant tissues continue to be characterized, approaches to target those aberrant pathways are being actively explored. The epidermal growth factor receptor (EGFR) is commonly overexpressed in NSCLC, particularly squamous cell carcinoma, and has been implicated in the development and progression of this disease, although a clear correlation with prognosis has not been established. Several different strategies have been developed to target and block the EGFR and its downstream effects, and some of them have been intensively studied in preclinical and clinical studies as a single-agent approach or in combination with radiation therapy or chemotherapy. In this article, we review the role of EGFR in lung cancer, as well as preclinical and clinical data on strategies to interfere with EGFR signaling alone or in combination with chemotherapy, radiation, or both.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/fisiologia , Neoplasias Pulmonares/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/fisiopatologia , Ensaios Clínicos como Assunto , Reparo do DNA , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/fisiopatologia , Prognóstico , Proteínas Tirosina Quinases
18.
Oncology (Williston Park) ; 17(5 Suppl 5): 15-24, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12800601

RESUMO

Radiation therapy has traditionally been the treatment of choice for locally or regionally advanced cancer, but its therapeutic efficacy is often hindered by limited tolerance of normal tissues and by tumor radioresistance. To improve therapeutic outcome, radiotherapy is frequently combined with chemotherapeutic drugs that are themselves cytotoxic and may sensitize cells to radiation. Solid evidence exists that administering standard chemotherapeutic agents during the course of radiotherapy (concurrent chemoradiotherapy) increases both local tumor control and patient survival in a number of cancer sites. These therapeutic improvements, however, have been achieved at the expense of considerable normal tissue toxicity. To improve chemoradiotherapy further, there have been extensive explorations of the potential of newer chemotherapeutic agents, including irinotecan (CPT-11, Camptosar) and other topoisomerase inhibitors. Preclinical studies have shown that these agents are potent radiosensitizers, providing a strong biologic rationale for using these drugs in combination with radiotherapy. These studies also generated information critical for designing effective treatment schedules in clinical settings. The therapeutic efficacy of topoisomerase inhibitor-radiation combinations is currently being tested clinically. Recent advances in molecular biology have discovered many cellular molecules, including the cyclooxygenase-2 (COX-2) enzyme, that promote tumor cell survival and are responsible for tumor resistance to cytotoxic agents, and hence may serve as potential targets for augmentation of radio (or chemo) response. COX-2 is often overexpressed in premalignant lesions and cancer, and is involved in carcinogenesis, tumor growth, and metastatic spread. Preclinical studies provided solid evidence that inhibition of this enzyme with selective COX-2 inhibitors prevents carcinogenesis, slows the growth of established tumors, and enhances tumor response to radiation without appreciably affecting normal tissue radioresponse. The mechanisms of enhancement of tumor radioresponse involve direct actions on tumor cells and indirect actions, primarily on tumor vasculature. COX-2 inhibitors also improve tumor response to chemotherapeutic agents, including irinotecan. Additional therapeutic benefit was observed for celecoxib (Celebrex), a selective COX-2 inhibitor, consisting of a strong reduction in irinotecan-induced diarrhea. Thus, selective targeting of COX-2 may potentially improve radiotherapy, chemotherapy, or chemoradiotherapy--a therapeutic strategy that is currently being tested in clinical trials.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Camptotecina/análogos & derivados , Isoenzimas/efeitos dos fármacos , Isoenzimas/efeitos da radiação , Prostaglandina-Endoperóxido Sintases/efeitos dos fármacos , Prostaglandina-Endoperóxido Sintases/efeitos da radiação , Radioterapia , Animais , Camptotecina/uso terapêutico , Ensaios Clínicos como Assunto , Terapia Combinada , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/uso terapêutico , Humanos , Irinotecano , Proteínas de Membrana , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Neoplasias/radioterapia
19.
Am J Clin Oncol ; 37(1): 90-100, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22495455

RESUMO

Recent research on inhibitors of poly (ADP-ribose) polymerase (PARP) has demonstrated their potential for improving cancer therapy. They inhibit protein poly (ADP-ribosyl)ation and thus affect numerous molecular and cellular functions, including DNA repair and cell survival, that are critical for such physiological and patho-physiological states as carcinogenesis, inflammation, and resistance to cancer therapy. In this review, we describe the biological basis underlying the use of these agents in cancer therapy, providing data from preclinical studies that demonstrate the synergistic interaction of PARP inhibitors with radiation and chemotherapeutics. We also summarize initial clinical trials of PARP inhibitors for cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Antineoplásicos/uso terapêutico , Apoptose , Ensaios Clínicos como Assunto , Reparo do DNA , Sinergismo Farmacológico , Humanos , Inflamação , Necrose , Neoplasias/radioterapia , Neovascularização Patológica , Poli(ADP-Ribose) Polimerases/metabolismo , Radioterapia Adjuvante
20.
Anticancer Res ; 34(12): 6981-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25503124

RESUMO

BACKGROUND: Positive transcription elongation factor-b (P-TEFb) is a complex containing CDK9 and a cyclin (T1, T2 or K). The effect of inhibition of P-TEFb by 5,6-dichloro-l-ß-D-ribofuranosyl benzimidazole (DRB) on cell radiosensitivity and the underlying mechanisms were investigated. MATERIALS AND METHODS: Six human cancer cell lines were subjected to (3)H-uridine incorporation, cell viability and clonogenic cell survival assays; cell-cycle redistribution and apoptosis assay; western blots and nuclear 53BP1 foci analysis after exposing the cells to DRB with/without γ-radiation. RESULTS: DRB suppressed colony formation and enhanced radiosensitivity of all cell lines. DRB caused a further increase in radiation-induced apoptosis and cell-cycle redistribution depending on p53 status. DRB prolonged the presence of radiation-induced nuclear p53 binding protein-1 (53BP1) foci and suppressed the expression of sirtuin-1 (SIRT1) and casein kinase 2-alpha (CK2α), suggesting an inhibition of DNA repair processes. CONCLUSION: Our findings indicate that DRB has the potential to increase the efficacy of radiotherapy and warrants further investigation using in vivo tumor models.


Assuntos
Caseína Quinase II/antagonistas & inibidores , Diclororribofuranosilbenzimidazol/farmacologia , Fator B de Elongação Transcricional Positiva/antagonistas & inibidores , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/farmacologia , Sirtuína 1/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Caseína Quinase II/biossíntese , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos da radiação , Sirtuína 1/biossíntese , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA