Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 232
Filter
1.
Mol Cancer Ther ; 23(4): 577-588, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38359816

ABSTRACT

Treatments involving radiation and chemotherapy alone or in combination have improved patient survival and quality of life. However, cancers frequently evade these therapies due to adaptation and tumor evolution. Given the complexity of predicting response based solely on the initial genetic profile of a patient, a predetermined treatment course may miss critical adaptation that can cause resistance or induce new targets for drug and immunotherapy. To address the timescale for these evasive mechanisms, using a mouse xenograft tumor model, we investigated the rapidity of gene expression (mRNA), molecular pathway, and phosphoproteome changes after radiation, an HSP90 inhibitor, or combination. Animals received radiation, drug, or combination treatment for 1 or 2 weeks and were then euthanized along with a time-matched untreated group for comparison. Changes in gene expression occur as early as 1 week after treatment initiation. Apoptosis and cell death pathways were activated in irradiated tumor samples. For the HSP90 inhibitor and combination treatment at weeks 1 and 2 compared with Control Day 1, gene-expression changes induced inhibition of pathways including invasion of cells, vasculogenesis, and viral infection among others. The combination group included both drug-alone and radiation-alone changes. Our data demonstrate the rapidity of gene expression and functional pathway changes in the evolving tumor as it responds to treatment. Discovering these phenotypic adaptations may help elucidate the challenges in using sustained treatment regimens and could also define evolving targets for therapeutic efficacy.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Humans , Heterografts , Multiomics , Quality of Life , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/radiotherapy , HSP90 Heat-Shock Proteins , Cell Line, Tumor , Xenograft Model Antitumor Assays
2.
Cell Death Differ ; 30(5): 1382-1396, 2023 05.
Article in English | MEDLINE | ID: mdl-37055579

ABSTRACT

TNFα is a key mediator of immune, chemotherapy and radiotherapy-induced cytotoxicity, but several cancers, including head and neck squamous cell carcinomas (HNSCC), display resistance to TNFα due to activation of the canonical NFκB pro-survival pathway. However, direct targeting of this pathway is associated with significant toxicity; thus, it is vital to identify novel mechanism(s) contributing to NFκB activation and TNFα resistance in cancer cells. Here, we demonstrate that the expression of proteasome-associated deubiquitinase USP14 is significantly increased in HNSCC and correlates with worse progression free survival in Human Papillomavirus (HPV)- HNSCC. Inhibition or depletion of USP14 inhibited the proliferation and survival of HNSCC cells. Further, USP14 inhibition reduced both basal and TNFα-inducible NFκB activity, NFκB-dependent gene expression and the nuclear translocation of the NFκB subunit RELA. Mechanistically, USP14 bound to both RELA and IκBα and reduced IκBα K48-ubiquitination leading to the degradation of IκBα, a critical inhibitor of the canonical NFκB pathway. Furthermore, we demonstrated that b-AP15, an inhibitor of USP14 and UCHL5, sensitized HNSCC cells to TNFα-mediated cell death, as well as radiation-induced cell death in vitro. Finally, b-AP15 delayed tumor growth and enhanced survival, both as a monotherapy and in combination with radiation, in HNSCC tumor xenograft models in vivo, which could be significantly attenuated by TNFα depletion. These data offer new insights into the activation of NFκB signaling in HNSCC and demonstrate that small molecule inhibitors targeting the ubiquitin pathway warrant further investigation as a novel therapeutic avenue to sensitize these cancers to TNFα- and radiation-induced cytotoxicity.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Humans , Squamous Cell Carcinoma of Head and Neck/drug therapy , NF-KappaB Inhibitor alpha/genetics , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/genetics , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , NF-kappa B , Cell Death , Cell Line, Tumor , Ubiquitin Thiolesterase/genetics
3.
Antioxid Redox Signal ; 39(7-9): 432-444, 2023 09.
Article in English | MEDLINE | ID: mdl-37051681

ABSTRACT

Aims: Pancreatic ductal adenocarcinomas (PDACs) form hypovascular and hypoxic tumors, which are difficult to treat with current chemotherapy regimens. Gemcitabine (GEM) is often used as a first-line treatment for PDACs but has issues with chemoresistance and penetration in the interior of the tumor. Evofosfamide, a hypoxia-activated prodrug, has been shown to be effective in combination with GEM, although the mechanism of each drug on the other has not been established. We used mouse xenografts from two cell lines (MIA Paca-2 and SU.86.86) with different tumor microenvironmental characteristics to probe the action of each drug on the other. Results: GEM treatment enhanced survival times in mice with SU.86.86 leg xenografts (hazard ratio [HR] = 0.35, p = 0.03) but had no effect on MIA Paca-2 mice (HR = 0.91, 95% confidence interval = 0.37-2.25, p = 0.84). Conversely, evofosfamide did not improve survival times in SU.86.86 mice to a statistically significant degree (HR = 0.57, p = 0.22). Electron paramagnetic resonance imaging showed that oxygenation worsened in MIA Paca-2 tumors when treated with GEM, providing a direct mechanism for the activation of the hypoxia-activated prodrug evofosfamide by GEM. Sublethal amounts of either treatment enhanced the toxicity of other treatment in vitro in SU.86.86 but not in MIA Paca-2. By the biomarker γH2AX, combination treatment increased the number of double-stranded DNA lesions in vitro for SU.86.86 but not MIA Paca-2. Innovation and Conclusion: The synergy between GEM and evofosfamide appears to stem from the dual action of GEMs effect on tumor vasculature and inhibition by GEM of the homologous recombination DNA repair process. Antioxid. Redox Signal. 39, 432-444.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Prodrugs , Humans , Animals , Mice , Gemcitabine , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Heterografts , Prodrugs/pharmacology , Prodrugs/therapeutic use , Recombinational DNA Repair , Cell Line, Tumor , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Hypoxia/drug therapy , Pancreatic Neoplasms
4.
J Am Chem Soc ; 145(11): 6257-6269, 2023 Mar 22.
Article in English | MEDLINE | ID: mdl-36893341

ABSTRACT

Redox intercalation involves coupled ion-electron motion within host materials, finding extensive application in energy storage, electrocatalysis, sensing, and optoelectronics. Monodisperse MOF nanocrystals, compared to their bulk phases, exhibit accelerated mass transport kinetics that promote redox intercalation inside nanoconfined pores. However, nanosizing MOFs significantly increases their external surface-to-volume ratios, making the intercalation redox chemistry into MOF nanocrystals difficult to understand due to the challenge of differentiating redox sites at the exterior of MOF particles from the internal nanoconfined pores. Here, we report that Fe(1,2,3-triazolate)2 possesses an intercalation-based redox process shifted ca. 1.2 V from redox at the particle surface. Such distinct chemical environments do not appear in idealized MOF crystal structures but become magnified in MOF nanoparticles. Quartz crystal microbalance and time-of-flight secondary ion mass spectrometry combined with electrochemical studies identify the existence of a distinct and highly reversible Fe2+/Fe3+ redox event occurring within the MOF interior. Systematic manipulation of experimental parameters (e.g., film thickness, electrolyte species, solvent, and reaction temperature) reveals that this feature arises from the nanoconfined (4.54 Å) pores gating the entry of charge-compensating anions. Due to the requirement for full desolvation and reorganization of electrolyte outside the MOF particle, the anion-coupled oxidation of internal Fe2+ sites involves a giant redox entropy change (i.e., 164 J K-1 mol-1). Taken together, this study establishes a microscopic picture of ion-intercalation redox chemistry in nanoconfined environments and demonstrates the synthetic possibility of tuning electrode potentials by over a volt, with profound implications for energy capture and storage technologies.

5.
Cancers (Basel) ; 15(4)2023 Feb 06.
Article in English | MEDLINE | ID: mdl-36831373

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) remains a prevalent diagnosis with current treatment options that include radiotherapy and immune-mediated therapies, in which tumor necrosis factor-α (TNFα) is a key mediator of cytotoxicity. However, HNSCC and other cancers often display TNFα resistance due to activation of the canonical IKK-NFκB/RELA pathway, which is activated by, and induces expression of, cellular inhibitors of apoptosis proteins (cIAPs). Our previous studies have demonstrated that the IAP inhibitor birinapant sensitized HNSCC to TNFα-dependent cell death in vitro and radiotherapy in vivo. Furthermore, we recently demonstrated that the inhibition of the G2/M checkpoint kinase WEE1 also sensitized HNSCC cells to TNFα-dependent cell death, due to the inhibition of the pro-survival IKK-NFκB/RELA complex. Given these observations, we hypothesized that dual-antagonist therapy targeting both IAP and WEE1 proteins may have the potential to synergistically sensitize HNSCC to TNFα-dependent cell death. Using the IAP inhibitor birinapant and the WEE1 inhibitor AZD1775, we show that combination treatment reduced cell viability, proliferation and survival when compared with individual treatment. Furthermore, combination treatment enhanced the sensitivity of HNSCC cells to TNFα-induced cytotoxicity via the induction of apoptosis and DNA damage. Additionally, birinapant and AZD1775 combination treatment decreased cell proliferation and survival in combination with radiotherapy, a critical source of TNFα. These results support further investigation of IAP and WEE1 inhibitor combinations in preclinical and clinical studies in HNSCC.

6.
J Radiat Res ; 64(1): 24-32, 2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36253079

ABSTRACT

Non-lethal doses of ionizing radiation (IR) delivered to humans because of terrorist events, nuclear accidents or radiotherapy can result in carcinogenesis. Means of protecting against carcinogenesis are lacking. We questioned the role of the gut microbiome in IR-induced carcinogenesis. The gut microbiome was modulated by administering broad spectrum antibiotics (Ab) in the drinking water. Mice were given Ab 3 weeks before and 3 weeks after 3 Gy total body irradiation (TBI) or for 6 weeks one month after TBI. Three weeks of Ab treatment resulted in a 98% reduction in total 16S rRNA counts for 4 out of 6 of the phylum groups detected. However, 3 more weeks of Ab treatment (6 weeks total) saw an expansion in the phylum groups Proteobacteria and Actinobacteria. The Ab treatment altered the bacteria diversity in the gut, and shortened the lifespan when Ab were administered before and after TBI. Mortality studies indicated that the adverse Ab lifespan effects were due to a decrease in the time in which solid tumors started to appear and not to any changes in hematopoietic or benign tumors. In contrast, when Ab were administered one month after TBI, lifespan was unchanged compared to the control TBI group. Use of broad-spectrum antibiotics to simulate the germ-free condition did not afford an advantage on carcinogenesis or lifespan.


Subject(s)
Gastrointestinal Microbiome , Humans , Mice , Animals , RNA, Ribosomal, 16S/genetics , Carcinogenesis , Whole-Body Irradiation/adverse effects , Anti-Bacterial Agents/pharmacology
7.
Neoplasia ; 30: 100793, 2022 08.
Article in English | MEDLINE | ID: mdl-35523073

ABSTRACT

PURPOSE: PEGylated human hyaluronidase (PEGPH20) enzymatically depletes hyaluronan, an important component of the extracellular matrix, increasing the delivery of therapeutic molecules. Combinations of chemotherapy and PEGPH20, however, have been unsuccessful in Phase III clinical trials. We hypothesize that by increasing tumor oxygenation by improving vascular patency and perfusion, PEGPH20 will also act as a radiosensitization agent. EXPERIMENTAL DESIGN: The effect of PEGPH20 on radiation treatment was analyzed with respect to tumor growth, survival time, p02, local blood volume, and the perfusion/permeability of blood vessels in a human pancreatic adenocarcinoma BxPC3 mouse model overexpressing hyaluronan synthase 3 (HAS3). RESULTS: Mice overexpressing HAS3 developed fast growing, radiation resistant tumors that became rapidly more hypoxic as time progressed. Treatment with PEGPH20 increased survival times when used in combination with radiation therapy, significantly more than either radiation therapy or PEGPH20 alone. In mice that overexpressed HAS3, EPR imaging showed an increase in local pO2 that could be linked to increases in perfusion/permeability and local blood volume immediately after PEGPH20 treatment. Hyperpolarized [1-13C] pyruvate suggested PEGPH20 caused a metabolic shift towards decreased glycolytic flux. These effects were confined to the mice overexpressing HAS3 - no effect of PEGPH20 on survival, radiation treatment, or pO2 was seen in wild type BxPC3 tumors. CONCLUSIONS: PEGPH20 may be useful for radiosensitization of pancreatic cancer but only in the subset of tumors with substantial hyaluronan accumulation. The response of the treatment may potentially be monitored by non-invasive imaging of the hemodynamic and metabolic changes in the tumor microenvironment.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Adenocarcinoma/drug therapy , Animals , Heterografts , Humans , Hyaluronic Acid/metabolism , Hyaluronic Acid/pharmacology , Hyaluronoglucosaminidase/metabolism , Hyaluronoglucosaminidase/pharmacology , Hyaluronoglucosaminidase/therapeutic use , Mice , Molecular Imaging , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/radiotherapy , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use , Tumor Microenvironment , Pancreatic Neoplasms
8.
Int J Oncol ; 60(6)2022 Jun.
Article in English | MEDLINE | ID: mdl-35438186

ABSTRACT

The purpose of the present trial was to determine the feasibility of the daily topical application of the piperidine nitroxide, MTS­01, combined with chemoradiotherapy in the treatment of patients with anal carcinoma. The secondary study endpoints were the description of the effects of this agent on skin toxicity and rectal­associated lymphoid tissue. The participants received radiotherapy concurrent with mitomycin­C and 5­fluorouracil for carcinoma of the anal canal. MTS­01 was applied to the bilateral inguinal area and the gluteal cleft. Dermatologic and non­dermatologic toxicity was graded throughout the treatment period. Circulating lymphocytes were serially collected for phenotyping. Rectal mucosal snag biopsies were collected at baseline and at 1 year of follow­up. A total of 5 patients received topical MTS­01. Adverse events attributed to MTS­01 included asymptomatic grade 1 hypoglycemia and grade 1­2 diarrhea. Dermatitis within untreated, radiated skin was not more severe than dermatitis in MTS­01­treated, unirradiated skin. Circulating CD4+ lymphocyte suppression was noted at >1 year following treatment in human immunodeficiency virus­negative participants. CD4+ lymphocytes remained suppressed in the irradiated rectal mucosa at 1 year, whereas the CD8+ lymphocyte numbers recovered or increased. On the whole, the present study demonstrates that the MTS­01 topical application was tolerable with minimal toxicity. Chemoradiation for anal cancer led to prolonged CD4+ lymphocytopenia in the circulation and gut mucosa.


Subject(s)
Anus Neoplasms , Carcinoma, Squamous Cell , Chemoradiotherapy , Dermatitis , Anal Canal/pathology , Anus Neoplasms/pathology , Anus Neoplasms/therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Chemoradiotherapy/adverse effects , Dermatitis/etiology , Dermatitis/prevention & control , Fluorouracil , Humans , Neoplasm Staging , Pilot Projects
9.
JCI Insight ; 7(5)2022 03 08.
Article in English | MEDLINE | ID: mdl-35260537

ABSTRACT

Therapeutic IL-12 has demonstrated the ability to reduce local immune suppression in preclinical models, but clinical development has been limited by severe inflammation-related adverse events with systemic administration. Here, we show that potent immunologic tumor control of established syngeneic carcinomas can be achieved by i.t. administration of a tumor-targeted IL-12 antibody fusion protein (NHS-rmIL-12) using sufficiently low doses to avoid systemic toxicity. Single-cell transcriptomic analysis and ex vivo functional assays of NHS-rmIL-12-treated tumors revealed reinvigoration and enhanced proliferation of exhausted CD8+ T lymphocytes, induction of Th1 immunity, and a decrease in Treg number and suppressive capacity. Similarly, myeloid cells transitioned toward inflammatory phenotypes and displayed reduced suppressive capacity. Cell type-specific IL-12 receptor-KO BM chimera studies revealed that therapeutic modulation of both lymphoid and myeloid cells is required for maximum treatment effect and tumor cure. Study of single-cell data sets from human head and neck carcinomas revealed IL-12 receptor expression patterns similar to those observed in murine tumors. These results describing the diverse mechanisms underlying tumor-directed IL-12-induced antitumor immunity provide the preclinical rationale for the clinical study of i.t. NHS-IL-12.


Subject(s)
Carcinoma , Interleukin-12 , Animals , CD8-Positive T-Lymphocytes , Interleukin-12/genetics , Interleukin-12/metabolism , Mice , Receptors, Interleukin-12/genetics , Receptors, Interleukin-12/metabolism , T-Lymphocytes, Regulatory
10.
Free Radic Biol Med ; 178: 380-390, 2022 01.
Article in English | MEDLINE | ID: mdl-34883252

ABSTRACT

The nitroxide, Tempol, prevents obesity related changes in mice fed a high fat diet (HFD). The purpose of this study was to gain insight into the mechanisms that result in such changes by Tempol in female C3H mice. Microarray methodology, Western blotting, bile acid analyses, and gut microbiome sequencing were used to identify multiple genes, proteins, bile acids, and bacteria that are regulated by Tempol in female C3H mice on HFD. The effects of antibiotics in combination with Tempol on the gut microflora were also studied. Adipose tissue, from Tempol treated mice, was analyzed using targeted gene microarrays revealing up-regulation of fatty acid metabolism genes (Acadm and Acadl > 4-fold, and Acsm3 and Acsm5 > 10-fold). Gene microarray studies of liver tissue from mice switched from HFD to Tempol HFD showed down-regulation of fatty acid synthesis genes and up-regulation of fatty acid oxidation genes. Analyses of proteins involved in obesity revealed that the expression of aldehyde dehydrogenase 1A1 (ALDH1A1) and fasting induced adipose factor/angiopoietin-like protein 4 (FIAF/ANGPTL4) was altered by Tempol HFD. Bile acid studies revealed increases in cholic acid (CA) and deoxycholic acid (DCA) in both the liver and serum of Tempol treated mice. Tempol HFD effect on the gut microbiome composition showed an increase in the population of Akkermansia muciniphila, a bacterial species known to be associated with a lean, anti-inflammatory phenotype. Antibiotic treatment significantly reduced the total level of bacterial numbers, however, Tempol was still effective in reducing the HFD weight gain. Even after antibiotic treatment Tempol still positively influenced several bacterial species such as as Akkermansia muciniphila and Bilophila wadsworthia. The positive effects of Tempol moderating weight gain in female mice fed a HFD involves changes to the gut microbiome, bile acids composition, and finally to changes in genes and proteins involved in fatty acid metabolism and storage.


Subject(s)
Diet, High-Fat , Gastrointestinal Microbiome , Animals , Antioxidants , Cyclic N-Oxides , Diet, High-Fat/adverse effects , Female , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Obesity/drug therapy , Spin Labels
11.
Int J Mol Sci ; 22(9)2021 May 03.
Article in English | MEDLINE | ID: mdl-34063570

ABSTRACT

Understanding the global metabolic changes during the senescence of tumor cells can have implications for developing effective anti-cancer treatment strategies. Ionizing radiation (IR) was used to induce senescence in a human colon cancer cell line HCT-116 to examine secretome and metabolome profiles. Control proliferating and senescent cancer cells (SCC) exhibited distinct morphological differences and expression of senescent markers. Enhanced secretion of pro-inflammatory chemokines and IL-1, anti-inflammatory IL-27, and TGF-ß1 was observed in SCC. Significantly reduced levels of VEGF-A indicated anti-angiogenic activities of SCC. Elevated levels of tissue inhibitors of matrix metalloproteinases from SCC support the maintenance of the extracellular matrix. Adenylate and guanylate energy charge levels and redox components NAD and NADP and glutathione were maintained at near optimal levels indicating the viability of SCC. Significant accumulation of pyruvate, lactate, and suppression of the TCA cycle in SCC indicated aerobic glycolysis as the predominant energy source for SCC. Levels of several key amino acids decreased significantly, suggesting augmented utilization for protein synthesis and for use as intermediates for energy metabolism in SCC. These observations may provide a better understanding of cellular senescence basic mechanisms in tumor tissues and provide opportunities to improve cancer treatment.


Subject(s)
Cellular Senescence/genetics , Colonic Neoplasms/genetics , Metabolic Networks and Pathways/genetics , Metabolome/genetics , Cellular Senescence/radiation effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HCT116 Cells , Humans , Interleukin-1/genetics , Interleukin-27/genetics , Metabolic Networks and Pathways/radiation effects , Metabolome/radiation effects , Radiation, Ionizing , Transforming Growth Factor beta1/genetics , Vascular Endothelial Growth Factor A/genetics
12.
Sci Rep ; 11(1): 12155, 2021 06 09.
Article in English | MEDLINE | ID: mdl-34108512

ABSTRACT

Drastic sensitivity enhancement of dynamic nuclear polarization is becoming an increasingly critical methodology to monitor real-time metabolic and physiological information in chemistry, biochemistry, and biomedicine. However, the limited number of available hyperpolarized 13C probes, which can effectively interrogate crucial metabolic activities, remains one of the major bottlenecks in this growing field. Here, we demonstrate [1-13C] N-acetyl cysteine (NAC) as a novel probe for hyperpolarized 13C MRI to monitor glutathione redox chemistry, which plays a central part of metabolic chemistry and strongly influences various therapies. NAC forms a disulfide bond in the presence of reduced glutathione, which generates a spectroscopically detectable product that is separated from the main peak by a 1.5 ppm shift. In vivo hyperpolarized MRI in mice revealed that NAC was broadly distributed throughout the body including the brain. Its biochemical transformation in two human pancreatic tumor cells in vitro and as xenografts differed depending on the individual cellular biochemical profile and microenvironment in vivo. Hyperpolarized NAC can be a promising non-invasive biomarker to monitor in vivo redox status and can be potentially translatable to clinical diagnosis.


Subject(s)
Acetylcysteine/metabolism , Brain/metabolism , Carbon Isotopes/analysis , Glutathione/metabolism , Pancreatic Neoplasms/pathology , Animals , Apoptosis , Cell Proliferation , Humans , Magnetic Resonance Imaging , Mice , Oxidation-Reduction , Pancreatic Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Cancer Res ; 81(13): 3693-3705, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33837042

ABSTRACT

Immune checkpoint blockade (ICB) has become a standard therapy for several cancers, however, the response to ICB is inconsistent and a method for noninvasive assessment has not been established to date. To investigate the capability of multimodal imaging to evaluate treatment response to ICB therapy, hyperpolarized 13C MRI using [1-13C] pyruvate and [1,4-13C2] fumarate and dynamic contrast enhanced (DCE) MRI was evaluated to detect early changes in tumor glycolysis, necrosis, and intratumor perfusion/permeability, respectively. Mouse tumor models served as platforms for high (MC38 colon adenocarcinoma) and low (B16-F10 melanoma) sensitivity to dual ICB of PD-L1 and CTLA4. Glycolytic flux significantly decreased following treatment only in the less sensitive B16-F10 tumors. Imaging [1,4-13C2] fumarate conversion to [1,4-13C2] malate showed a significant increase in necrotic cell death following treatment in the ICB-sensitive MC38 tumors, with essentially no change in B16-F10 tumors. DCE-MRI showed significantly increased perfusion/permeability in MC38-treated tumors, whereas a similar, but statistically nonsignificant, trend was observed in B16-F10 tumors. When tumor volume was also taken into consideration, each imaging biomarker was linearly correlated with future survival in both models. These results suggest that hyperpolarized 13C MRI and DCE MRI may serve as useful noninvasive imaging markers to detect early response to ICB therapy. SIGNIFICANCE: Hyperpolarized 13C MRI and dynamic contrast enhanced MRI in murine tumor models provide useful insight into evaluating early response to immune checkpoint blockade therapy.See related commentary by Cullen and Keshari, p. 3444.


Subject(s)
Colonic Neoplasms/pathology , Glycolysis , Immune Checkpoint Inhibitors/pharmacology , Magnetic Resonance Imaging/methods , Melanoma, Experimental/pathology , Molecular Imaging/methods , Pyruvic Acid/metabolism , Animals , Apoptosis , Cell Proliferation , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Female , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Int J Radiat Oncol Biol Phys ; 110(5): 1295-1305, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33838214

ABSTRACT

PURPOSE: Recent preclinical studies suggest combining the HSP90 inhibitor AT13387 (Onalespib) with radiation (IR) against colon cancer and head and neck squamous cell carcinoma (HNSCC). These studies emphasized that AT13387 downregulates HSP90 client proteins involved in oncogenic signaling and DNA repair mechanisms as major drivers of enhanced radiosensitivity. Given the large array of client proteins HSP90 directs, we hypothesized that other key proteins or signaling pathways may be inhibited by AT13387 and contribute to enhanced radiosensitivity. Metabolomic analysis of HSP90 inhibition by AT13387 was conducted to identify metabolic biomarkers of radiosensitization and whether modulations of key proteins were involved in IR-induced tumor vasculogenesis, a process involved in tumor recurrence. METHODS AND MATERIALS: HNSCC and non-small cell lung cancer cell lines were used to evaluate the AT13387 radiosensitization effect in vitro and in vivo. Flow cytometry, immunofluorescence, and immunoblot analysis were used to evaluate cell cycle changes and HSP90 client protein's role in DNA damage repair. Metabolic analysis was performed using liquid chromatography-Mass spectrometry. Immunohistochemical examination of resected tumors post-AT13387 and IR treatment were conducted to identify biomarkers of IR-induced tumor vasculogenesis. RESULTS: In agreement with recent studies, AT13387 treatment combined with IR resulted in a G2/M cell cycle arrest and inhibited DNA repair. Metabolomic profiling indicated a decrease in key metabolites in glycolysis and tricarboxylic acid cycle by AT13387, a reduction in Adenosine 5'-triphosphate levels, and rate-limiting metabolites in nucleotide metabolism, namely phosphoribosyl diphosphate and aspartate. HNSCC xenografts treated with the combination exhibited increased tumor regrowth delay, decreased tumor infiltration of CD45 and CD11b+ bone marrow-derived cells, and inhibition of HIF-1 and SDF-1 expression, thereby inhibiting IR-induced vasculogenesis. CONCLUSIONS: AT13387 treatment resulted in pharmacologic inhibition of cancer cell metabolism that was linked to DNA damage repair. AT13387 combined with IR inhibited IR-induced vasculogenesis, a process involved in tumor recurrence postradiotherapy. Combining AT13387 with IR warrants consideration of clinical trial assessment.


Subject(s)
Benzamides/pharmacology , DNA Repair , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Head and Neck Neoplasms/radiotherapy , Isoindoles/pharmacology , Radiation Tolerance/drug effects , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Animals , Aspartic Acid/pharmacology , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Colonic Neoplasms/radiotherapy , DNA Damage , DNA Repair/drug effects , DNA Repair/radiation effects , Down-Regulation , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/radiation effects , HSP90 Heat-Shock Proteins/metabolism , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Lung Neoplasms/radiotherapy , M Phase Cell Cycle Checkpoints/drug effects , M Phase Cell Cycle Checkpoints/radiation effects , Metabolomics , Mice , Mice, Nude , Neoplasm Recurrence, Local , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/prevention & control , Nucleotides/biosynthesis , Nucleotides/metabolism , Radiation Tolerance/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Xenograft Model Antitumor Assays
15.
Molecules ; 26(6)2021 Mar 14.
Article in English | MEDLINE | ID: mdl-33799481

ABSTRACT

Radiation therapy is one of the main modalities to treat cancer/tumor. The response to radiation therapy, however, can be influenced by physiological and/or pathological conditions in the target tissues, especially by the low partial oxygen pressure and altered redox status in cancer/tumor tissues. Visualizing such cancer/tumor patho-physiological microenvironment would be a useful not only for planning radiotherapy but also to detect cancer/tumor in an earlier stage. Tumor hypoxia could be sensed by positron emission tomography (PET), electron paramagnetic resonance (EPR) oxygen mapping, and in vivo dynamic nuclear polarization (DNP) MRI. Tissue oxygenation could be visualized on a real-time basis by blood oxygen level dependent (BOLD) and/or tissue oxygen level dependent (TOLD) MRI signal. EPR imaging (EPRI) and/or T1-weighted MRI techniques can visualize tissue redox status non-invasively based on paramagnetic and diamagnetic conversions of nitroxyl radical contrast agent. 13C-DNP MRI can visualize glycometabolism of tumor/cancer tissues. Accurate co-registration of those multimodal images could make mechanisms of drug and/or relation of resulted biological effects clear. A multimodal instrument, such as PET-MRI, may have another possibility to link multiple functions. Functional imaging techniques individually developed to date have been converged on the concept of theranostics.


Subject(s)
Electron Spin Resonance Spectroscopy/methods , Magnetic Resonance Imaging/methods , Neoplasms/pathology , Positron-Emission Tomography/methods , Tumor Microenvironment/physiology , Animals , Contrast Media/chemistry , Humans , Nitrogen Oxides/chemistry , Oxidation-Reduction , Oxygen/chemistry
16.
Antioxid Redox Signal ; 35(11): 904-915, 2021 10 10.
Article in English | MEDLINE | ID: mdl-32787454

ABSTRACT

Aims: In hypoxic tumor microenvironments, the strongly reducing redox environment reduces evofosfamide (TH-302) to release a cytotoxic bromo-isophosphoramide (Br-IPM) moiety. This drug therefore preferentially attacks hypoxic regions in tumors where other standard anticancer treatments such as chemotherapy and radiation therapy are often ineffective. Various combination therapies with evofosfamide have been proposed and tested in preclinical and clinical settings. However, the treatment effect of evofosfamide monotherapy on tumor hypoxia has not been fully understood, partly due to the lack of quantitative methods to assess tumor pO2in vivo. Here, we use quantitative pO2 imaging by electron paramagnetic resonance (EPR) to evaluate the change in tumor hypoxia in response to evofosfamide treatment using two pancreatic ductal adenocarcinoma xenograft models: MIA Paca-2 tumors responding to evofosfamide and Su.86.86 tumors that do not respond. Results: EPR imaging showed that oxygenation improved globally after evofosfamide treatment in hypoxic MIA Paca-2 tumors, in agreement with the ex vivo results obtained from hypoxia staining by pimonidazole and in apparent contrast to the decrease in Ktrans observed in dynamic contrast-enhanced magnetic resonance imaging (DCE MRI). Innovations: The observation that evofosfamide not only kills the hypoxic region of the tumor but also improves oxygenation in the residual tumor regions provides a rationale for combination therapies using radiation and antiproliferatives post evofosfamide for improved outcomes. Conclusion: This study suggests that reoxygenation after evofosfamide treatment is due to decreased oxygen demand rather than improved perfusion. Following the change in pO2 after treatment may therefore yield a way of monitoring treatment response. Antioxid. Redox Signal. 35, 904-915.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/therapy , Cell Hypoxia/drug effects , Nitroimidazoles/pharmacology , Pancreatic Neoplasms/therapy , Phosphoramide Mustards/pharmacology , Prodrugs/pharmacology , Animals , Antineoplastic Agents/chemistry , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , Mice , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Nitroimidazoles/chemistry , Oxidation-Reduction , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphoramide Mustards/chemistry , Prodrugs/chemistry
17.
Clin Cancer Res ; 26(22): 5781-5790, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32554542

ABSTRACT

Radiotherapy is highly effective due to its ability to physically focus the treatment to target the tumor while sparing normal tissue and its ability to be combined with systemic therapy. This systemic therapy can be utilized before radiotherapy as an adjuvant or induction treatment, during radiotherapy as a radiation "sensitizer," or following radiotherapy as a part of combined modality therapy. As part of a unique concept of using radiation as "focused biology," we investigated how tumors and normal tissues adapt to clinically relevant multifraction (MF) and single-dose (SD) radiation to observe whether the adaptations can induce susceptibility to cell killing by available drugs or by immune enhancement. We identified an adaptation occurring after MF (3 × 2 Gy) that induced cell killing when AKT-mTOR inhibitors were delivered following cessation of radiotherapy. In addition, we identified inducible changes in integrin expression 2 months following cessation of radiotherapy that differ between MF (1 Gy × 10) and SD (10 Gy) that remain targetable compared with preradiotherapy. Adaptation is reflected across different "omics" studies, and thus the range of possible molecular targets is not only broad but also time, dose, and schedule dependent. While much remains to be studied about the radiation adaptive response, radiation should be characterized by its molecular perturbations in addition to physical dose. Consideration of the adaptive effects should result in the design of a tailored radiotherapy treatment plan that accounts for specific molecular changes to be targeted as part of precision multimodality cancer treatment.


Subject(s)
Neoplasms/drug therapy , Neoplasms/radiotherapy , Radiation Injuries/drug therapy , Radiation Injuries/radiotherapy , Combined Modality Therapy , Dose Fractionation, Radiation , Humans , Immunotherapy , Neoplasms/genetics , Neoplasms/pathology , Oncogene Protein v-akt/antagonists & inhibitors , Radiation Injuries/pathology , Radiotherapy Dosage , TOR Serine-Threonine Kinases/antagonists & inhibitors
18.
Chem Rev ; 120(14): 6738-6782, 2020 Jul 22.
Article in English | MEDLINE | ID: mdl-32597172

ABSTRACT

There is an urgent global need for electrochemical energy storage that includes materials that can provide simultaneous high power and high energy density. One strategy to achieve this goal is with pseudocapacitive materials that take advantage of reversible surface or near-surface Faradaic reactions to store charge. This allows them to surpass the capacity limitations of electrical double-layer capacitors and the mass transfer limitations of batteries. The past decade has seen tremendous growth in the understanding of pseudocapacitance as well as materials that exhibit this phenomenon. The purpose of this Review is to examine the fundamental development of the concept of pseudocapacitance and how it came to prominence in electrochemical energy storage as well as to describe new classes of materials whose electrochemical energy storage behavior can be described as pseudocapacitive.

19.
Cancer Res ; 80(11): 2087-2093, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32245793

ABSTRACT

Molecular imaging approaches for metabolic and physiologic imaging of tumors have become important for treatment planning and response monitoring. However, the relationship between the physiologic and metabolic aspects of tumors is not fully understood. Here, we developed new hyperpolarized MRI and electron paramagnetic resonance imaging procedures that allow more direct assessment of tumor glycolysis and oxygenation status quantitatively. We investigated the spatial relationship between hypoxia, glucose uptake, and glycolysis in three human pancreatic ductal adenocarcinoma tumor xenografts with differing physiologic and metabolic characteristics. At the bulk tumor level, there was a strong positive correlation between 18F-FDG-PET and lactate production, while pO2 was inversely related to lactate production and 18F-2-fluoro-2-deoxy-D-glucose (18F-FDG) uptake. However, metabolism was not uniform throughout the tumors, and the whole tumor results masked different localizations that became apparent while imaging. 18F-FDG uptake negatively correlated with pO2 in the center of the tumor and positively correlated with pO2 on the periphery. In contrast to pO2 and 18F-FDG uptake, lactate dehydrogenase activity was distributed relatively evenly throughout the tumor. The heterogeneity revealed by each measure suggests a multimodal molecular imaging approach can improve tumor characterization, potentially leading to better prognostics in cancer treatment. SIGNIFICANCE: Novel multimodal molecular imaging techniques reveal the potential of three interrelated imaging biomarkers to profile the tumor microenvironment and interrelationships of hypoxia, glucose uptake, and glycolysis.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Glucose/metabolism , Oxygen/metabolism , Pancreatic Neoplasms/metabolism , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/diagnostic imaging , Cell Line, Tumor , Electron Spin Resonance Spectroscopy/methods , Fluorodeoxyglucose F18 , Glycolysis , Heterografts , Humans , Mice , Molecular Imaging/methods , Pancreatic Neoplasms/diagnostic imaging , Partial Pressure , Positron-Emission Tomography/methods , Radiopharmaceuticals , Tumor Microenvironment
20.
Cell Rep ; 30(6): 1798-1810.e4, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32049011

ABSTRACT

The reliance of many cancers on aerobic glycolysis has stimulated efforts to develop lactate dehydrogenase (LDH) inhibitors. However, despite significant efforts, LDH inhibitors (LDHi) with sufficient specificity and in vivo activity to determine whether LDH is a feasible drug target are lacking. We describe an LDHi with potent, on-target, in vivo activity. Using hyperpolarized magnetic resonance spectroscopic imaging (HP-MRSI), we demonstrate in vivo LDH inhibition in two glycolytic cancer models, MIA PaCa-2 and HT29, and we correlate depth and duration of LDH inhibition with direct anti-tumor activity. HP-MRSI also reveals a metabolic rewiring that occurs in vivo within 30 min of LDH inhibition, wherein pyruvate in a tumor is redirected toward mitochondrial metabolism. Using HP-MRSI, we show that inhibition of mitochondrial complex 1 rapidly redirects tumor pyruvate toward lactate. Inhibition of both mitochondrial complex 1 and LDH suppresses metabolic plasticity, causing metabolic quiescence in vitro and tumor growth inhibition in vivo.


Subject(s)
Drug Therapy, Combination/methods , L-Lactate Dehydrogenase/antagonists & inhibitors , Neoplasms/immunology , Animals , Humans , Mice , Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...