Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Int J Mol Sci ; 24(13)2023 Jun 25.
Article in English | MEDLINE | ID: mdl-37445799

ABSTRACT

Current treatment for prostate cancer is dependent on the stages of the cancer, recurrence, and genetic factors. Treatment varies from active surveillance or watchful waiting to prostatectomy, chemotherapy, and radiation therapy in combination or alone. Although radical prostate cancer therapy reduces the advancement of the disease and its mortality, the increased disease treatment associated morbidity, erectile dysfunction, and incontinence affect the quality of life of cancer survivors. To overcome these problems, photodynamic therapy (PDT) has previously been investigated using PhotofrinTM as a photosensitizer (PS). However, Photofrin-PDT has shown limitations in treating prostate cancer due to its limited tumor-specificity and the depth of light penetration at 630 nm (the longest wavelength absorption of PhotofrinTM). The results presented herein show that this limitation can be solved by using a near infrared (NIR) compound as a photosensitizer (PS) for PDT and the same agent also acts as a sonosensitizer for SDT (using ultrasound to activate the compound). Compared to light, ultrasound has a stronger penetration ability in biological tissues. Exposing the PS (or sonosensitizer) to ultrasound (US) initiates an electron-transfer process with a biological substrate to form radicals and radical ions (type I reaction). In contrast, exposure of the PS to light (PDT) generates singlet oxygen (type II reaction). Therefore, the reactive oxygen species (ROS) produced by SDT and PDT follow two distinct pathways, i.e., type I (oxygen independent) and type II (oxygen dependent), respectively, and results in significantly enhanced destruction of tumor cells. The preliminary in vitro and in vivo results in a PC3 cell line and tumor model indicate that the tumor specificality of the therapeutic agent(s) can be increased by targeting galectin-1 and galectin-3, known for their overexpression in prostate cancer.


Subject(s)
Photochemotherapy , Prostatic Neoplasms , Male , Humans , Mice , Animals , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/chemistry , Photochemotherapy/methods , Dihematoporphyrin Ether , Quality of Life , Prostatic Neoplasms/pathology , Oxygen , Cell Line, Tumor
2.
Molecules ; 28(9)2023 Apr 27.
Article in English | MEDLINE | ID: mdl-37175191

ABSTRACT

To investigate and compare the pharmacokinetic profile and anti-cancer activity of fluorinated and iodinated photosensitizers (PSs), the 3-(1'-(o-fluorobenzyloxy)ethyl pyropheophorbide and the corresponding meta-(m-) and para (p-) fluorinated analogs (methyl esters and carboxylic acids) were synthesized. Replacing iodine with fluorine in PSs did not make any significant difference in fluorescence and singlet oxygen (a key cytotoxic agent) production. The nature of the delivery vehicle and tumor types showed a significant difference in uptake and long-term cure by photodynamic therapy (PDT), especially in the iodinated PS. An unexpected difference in the pharmacokinetic profiles of fluorinated vs. iodinated PSs was observed. At the same imaging parameters, the fluorinated PSs showed maximal tumor uptake at 2 h post injection of the PS, whereas the iodinated PS gave the highest uptake at 24 h post injection. Among all isomers, the m-fluoro PS showed the best in vivo anti-cancer activity in mice bearing U87 (brain) or bladder (UMUC3) tumors. A direct correlation between the tumor uptake and PDT efficacy was observed. The higher tumor uptake of m-fluoro PS at two hours post injection provides a solid rationale for developing the corresponding 18F-agent (half-life 110 min only) for positron imaging tomography (PET) of those cancers (e.g., bladder, prostate, kidney, pancreas, and brain) where 18F-FDG-PET shows limitations.


Subject(s)
Neoplasms , Photochemotherapy , Male , Animals , Mice , Photosensitizing Agents/therapeutic use , Chlorophyll A , Photochemotherapy/methods , Neoplasms/drug therapy , Chlorophyll/pharmacology , Cell Line, Tumor
3.
J Surg Res ; 263: 245-250, 2021 07.
Article in English | MEDLINE | ID: mdl-33713956

ABSTRACT

BACKGROUND: Lung cancer is the greatest cause of cancer mortality in the United States, necessitating ongoing improvements in current treatment techniques. Photodynamic therapy (PDT) involves the interaction between a photosensitizer, light, and oxygen. The resulting release of reactive oxygen species causes tumor necrosis. It has been used as an endoscopic technique for the palliation of lung cancer. Porfimer sodium (Photofrin) is the only Food and Drug Administration-approved photosensitizer for PDT but has limited depth of penetration and produces prolonged skin phototoxicity. Multiple newer photosensitizers are in development, including PS785. The effectiveness of PS785 was compared with porfimer sodium in the treatment of human lung cancer xenografts in mice. METHODS: Human non-small cell lung cancer (NSCLC) xenografts were established in severe combined immunodeficient mice and grouped into small (3-5 mm) and large tumors (6-10 mm). PS785 or porfimer sodium was administered intravenously, and PDT was executed at 24, 48, or 72 h after injection. The primary endpoint was the delay of tumor regrowth after PDT. RESULTS: Porfimer sodium and PS785 produced statistically similar delays of tumor regrowth after PDT when small tumors were treated at 24 and 48 h. At 72 h, PS785 performed better in small tumors. However, for large tumors, PS785 produced no delay in tumor regrowth at any time point. CONCLUSIONS: PS785 and porfimer sodium were able to effectively treat NSCLC to a depth of ≤5 mm. However, porfimer sodium was more effective in treating NSCLC tumors to a depth of 6-10 mm. Further efforts are required to produce photosensitizers that will facilitate PDT of larger tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Dihematoporphyrin Ether/administration & dosage , Lung Neoplasms/therapy , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Injections, Intravenous , Lung/pathology , Lung/surgery , Lung Neoplasms/pathology , Mice , Pneumonectomy , Xenograft Model Antitumor Assays
4.
Lasers Surg Med ; 50(5): 506-512, 2018 07.
Article in English | MEDLINE | ID: mdl-29737551

ABSTRACT

BACKGROUND AND OBJECTIVES: In this study, we evaluated the impact of hyperthermia in photosensitizing efficacy of 3-[(1'-hexyloxy)ethyl-3-devinylpyropheophorbide-a (HPPH or Photochlor) for the treatment of cancer by photodynamic therapy (PDT). STUDY DESIGN/MATERIALS AND METHODS: The outcome of both whole body hyperthermia (WBH) and local hyperthermia (LH) in combination with HPPH-PDT was determined in BALB/c and nude mice bearing Colon26 and U87 tumors, respectively. LH was performed by using an indigenously designed heating device, that was heated to the required temperature using a circulating water bath. The device which has flexible membrane on one side was placed on skin above the tumor. The temperature of the tumor was monitored using a thermocouple sensor placed on the surface of the tumor capable of measuring the temperature within 0.1°C. Uptake of the photosensitizer in tumors was determined by fluorescence using an IVIS or a Nuance Imaging System. The PDT was performed by exposing the tumors to 665 nm laser loght, (135 J/cm2 , 75 mW/cm2 ) at the maximal uptake time of HPPH. Tumor size was measured daily using vernier calipers. RESULTS: The improved PDT efficacy (long-term percentage tumor cure) in combination with hyperthermia is possible due to an increase in tumor-uptake of the photosensitizer (PS), confirmed by in vivo fluorescence imaging and also by increased tumor perfusion and decreased hypoxia as have been reported previously (Sen et al. [2011] Cancer Res. 71:3872-3880 In Vivo. 20:689-695). Interestingly, compared to whole body hyperthermia, the 14 C- HPPH biodistribution data under local hyperthermia showed similar tumor-uptake in BALB/c mice bearing Colon26 tumors, but significantly lower uptake in other organs and in the blood. CONCLUSION: Our study demonstrates that both, fever range whole body and local hyperthermia in combination with HPPH-PDT enhances the long-term tumor cure of BALB/c and nude mice implanted with Colon26 and U87 tumors respectively. Lasers Surg. Med. 50:506-512, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Chlorophyll/analogs & derivatives , Colonic Neoplasms/therapy , Hyperthermia, Induced/methods , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Animals , Chlorophyll/pharmacology , Colonic Neoplasms/pathology , Disease Models, Animal , Mice , Mice, Inbred BALB C , Mice, Nude
5.
Nanomedicine ; 14(7): 2295-2305, 2018 10.
Article in English | MEDLINE | ID: mdl-30059754

ABSTRACT

Advances in in vivo stability and preferential tumor uptake of cancer nanomedicine are warranted for effective chemotherapy. Here, we describe a novel nanoformulation using an unconventional polymeric tubule-forming phospholipid, DC8,9PC. We report that DC8,9PC transitions to stable vesicles (LNPs) in the presence of PEGylated lipid (DSPE-PEG2000); the resulting DC8,9PC:DSPE-PEG2000 LNPs efficiently included a hydrophobic PDT drug, HPPH. Remarkably, these LNPs incorporated unusually high DSPE-PEG2000 concentrations; LNP10-HPPH and LNP20-HPPH (10 & 20 mol% PEGylated lipid, respectively) exhibited >90% serum stability at 37 °C. Increased PEGylation in the LNPs correlated with enhanced tumor accumulation in intravenously injected HT29 tumor mouse xenographs. Colon-26 bearing BALB/c mice, intravenously injected with LNP20-HPPH showed superior PDT efficacy and animal survival (no tumor recurrence up to 100 days) as compared to a formulation currently used in clinical trials. Taken together, we present a simple stealth binary lipid nanosystem with enhanced efficiency of tumor accumulation and superior therapeutic efficacy.


Subject(s)
Colorectal Neoplasms/drug therapy , Drug Delivery Systems , Nanoparticles/administration & dosage , Phospholipids/chemistry , Photochemotherapy , Photosensitizing Agents/administration & dosage , Polymers/chemistry , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Drug Carriers/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Photosensitizing Agents/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Int J Mol Sci ; 19(11)2018 Oct 29.
Article in English | MEDLINE | ID: mdl-30380599

ABSTRACT

Durable response, inherent or acquired resistance, and dose-limiting toxicities continue to represent major barriers in the treatment of patients with advanced clear-cell renal cell carcinoma (ccRCC). The majority of ccRCC tumors are characterized by the loss of Von Hippel⁻Lindau tumor suppressor gene function, a stable expression of hypoxia-inducible factors 1α and 2α (HIFs), an altered expression of tumor-specific oncogenic microRNAs (miRNAs), a clear cytoplasm with dense lipid content, and overexpression of thymidine phosphorylase. The aim of this manuscript was to confirm that the downregulation of specific drug-resistant biomarkers deregulated in tumor cells by a defined dose and schedule of methylselenocysteine (MSC) or seleno-l-methionine (SLM) sensitizes tumor cells to mechanism-based drug combination. The inhibition of HIFs by selenium was necessary for optimal therapeutic benefit. Durable responses were achieved only when MSC was combined with sunitinib (a vascular endothelial growth factor receptor (VEGFR)-targeted biologic), topotecan (a topoisomerase 1 poison and HIF synthesis inhibitor), and S-1 (a 5-fluorouracil prodrug). The documented synergy was selenium dose- and schedule-dependent and associated with enhanced prolyl hydroxylase-dependent HIF degradation, stabilization of tumor vasculature, downregulation of 28 oncogenic miRNAs, as well as the upregulation of 12 tumor suppressor miRNAs. The preclinical results generated provided the rationale for the development of phase 1/2 clinical trials of SLM in sequential combination with axitinib in ccRCC patients refractory to standard therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Renal Cell/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Kidney Neoplasms/drug therapy , MicroRNAs/genetics , Selenocysteine/analogs & derivatives , Selenomethionine/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Female , Fluorouracil/therapeutic use , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney Neoplasms/blood supply , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mice, Nude , Selenocysteine/therapeutic use , Topotecan/therapeutic use
7.
Biomed Pharmacother ; 168: 115731, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37857248

ABSTRACT

Photobac is a near infrared photosensitizer (PS) derived from naturally occurring bacteriochlorophyll- a, with a potential for treating a variety of cancer types (U87, F98 and C6 tumor cells in vitro). The main objective of the studies presented herein was to evaluate the efficacy, toxicity and pharmacokinetic profile of Photobac in animals (mice, rats and dogs) and submit these results to the United States Food and Drug Administration (US FDA) for its approval to initiate Phase I human clinical trials of glioblastoma, a deadly cancer disease with no long term cure. The photodynamic therapy (PDT) efficacy of Photobac was evaluated in mice subcutaneously implanted with U87 tumors, and in rats bearing C6 tumors implanted in brain. In both tumor types, the Photobac-PDT was quite effective. The long-term cure in rats was monitored by magnetic resonance imaging (MRI) and histopathology analysis. A detailed pharmacology, pharmacokinetics and toxicokinetic study of Photobac was investigated in both non-GLP and GLP facilities at variable doses following the US FDA parameters. Safety Pharmacology studies suggest that there is no phototoxicity, cerebral or retinal toxicity with Photobac. No metabolites of Photobac were observed following incubation in rat, dog, mini-pig and human hepatocytes. Based on current biological data, Photobac-IND received the approval for Phase-I human clinical trials to treat Glioblastoma (brain cancer), which is currently underway at our institute. Photobac has also received an orphan drug status from the US FDA, because of its potential for treating Glioblastoma as no effective treatment is currently available for this deadly disease.


Subject(s)
Brain Neoplasms , Glioblastoma , Photochemotherapy , Rats , Dogs , Animals , Mice , Humans , Swine , Bacteriochlorophylls/therapeutic use , Glioblastoma/pathology , Photochemotherapy/methods , Bacteriochlorophyll A/therapeutic use , Swine, Miniature , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Models, Animal
8.
BMC Cancer ; 12: 293, 2012 Jul 17.
Article in English | MEDLINE | ID: mdl-22804960

ABSTRACT

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) accounts for more than 80% of the cases of renal cell carcinoma. In ccRCC deactivation of Von-Hippel-Lindau (VHL) gene contributes to the constitutive expression of hypoxia inducible factors 1 and 2 alpha (HIF-α), transcriptional regulators of several genes involved in tumor angiogenesis, glycolysis and drug resistance. We have demonstrated inhibition of HIF-1α by Se-Methylselenocysteine (MSC) via stabilization of prolyl hydroxylases 2 and 3 (PHDs) and a significant therapeutic synergy when combined with chemotherapy. This study was initiated to investigate the expression of PHDs, HIF-α, and VEGF-A in selected solid cancers, the mechanism of HIF-α inhibition by MSC, and to document antitumor activity of MSC against human ccRCC xenografts. METHODS: Tissue microarrays of primary human cancer specimens (ccRCC, head & neck and colon) were utilized to determine the incidence of PHD2/3, HIF-α, and VEGF-A by immunohistochemical methods. To investigate the mechanism(s) of HIF-α inhibition by MSC, VHL mutated ccRCC cells RC2 (HIF-1α positive), 786-0 (HIF-2α positive) and VHL wild type head & neck cancer cells FaDu (HIF-1α) were utilized. PHD2 and VHL gene specific siRNA knockdown and inhibitors of PHD2 and proteasome were used to determine their role in the degradation of HIF-1α by MSC. RESULTS: We have demonstrated that ccRCC cells express low incidence of PHD2 (32%), undetectable PHD3, high incidence of HIF-α (92%), and low incidence of VEGF-A compared to head & neck and colon cancers. This laboratory was the first to identify MSC as a highly effective inhibitor of constitutively expressed HIF-α in ccRCC tumors. MSC did not inhibit HIF-1α protein synthesis, but facilitated its degradation. The use of gene knockdown and specific inhibitors confirmed that the inhibition of HIF-1α was PHD2 and proteasome dependent and VHL independent. The effects of MSC treatment on HIF-α were associated with significant antitumor activity against ccRCC xenograft. CONCLUSIONS: Our results show the role of PHD2/3 in stable expression of HIF-α in human ccRCC. Furthermore, HIF-1α degradation by MSC is achieved through PHD2 dependent and VHL independent pathway which is unique for HIF-α regulation. These data provide the basis for combining MSC with currently used agents for ccRCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/metabolism , Organoselenium Compounds/pharmacology , Procollagen-Proline Dioxygenase/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Dioxygenases/genetics , Dioxygenases/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases , Kidney Neoplasms/genetics , Mice , Mice, Nude , Procollagen-Proline Dioxygenase/genetics , Protein Processing, Post-Translational/drug effects , Proteolysis/drug effects , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
9.
Biomedicines ; 10(4)2022 Apr 06.
Article in English | MEDLINE | ID: mdl-35453607

ABSTRACT

We have previously shown that a radioactive (123I)-analog of methyl 3-(1'-(iodobexyloxy) ethyl-3-devinylpyropheophorbide-a (PET-ONCO), derived from chlorophyll-a can be used for positron emission tomography (PET) imaging of a variety of tumors, including those where 18F-FDG shows limitations. In this study, the photodynamic therapy (PDT) efficacy of the corresponding non-radioactive photosensitizer (PS) was investigated in a variety of tumor types (NSCLC, SCC, adenocarcinoma) derived from lung cancer patients in mice tumor models. The in vitro and in vivo efficacy was also investigated in combination with doxorubicin, and a significantly enhanced long-term tumor response was observed. The toxicity and toxicokinetic profile of the iodinated PS was also evaluated in male and female Sprague-Dawley rats and Beagle dog at variable doses (single intravenous injections) to assess reversibility or latency of any effects over a 28-day dose free period. The no-observed-adverse-effect (NOAEL) of the PS was considered to be 6.5 mg/kg for male and female rats, and for dogs, 3.45 mg/kg, the highest dose levels evaluated, respectively. The corresponding plasma Cmax and AYClast for male and female rats were 214,000 and 229,000 ng/mL and 3,680,000 and 3,810,000 h * ng/mL, respectively. For male and female dogs, the corresponding plasma Cmax and AYClast were 76,000 and 92,400 ng/mL and 976,000 and 1,200,000 h * ng/mL, respectively.

10.
J Med Chem ; 65(13): 9267-9280, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35763292

ABSTRACT

3-(1'-Hexyloxyethyl)-3-devinylpyropheophorbide-a (HPPH or Photochlor), a tumor-avid chlorophyll a derivative currently undergoing human clinical trials, was conjugated with mono-, di-, and tri-Gd(III)tetraxetan (DOTA) moieties. The T1/T2 relaxivity and in vitro PDT efficacy of these conjugates were determined. The tumor specificity of the most promising conjugate was also investigated at various time points in mice and rats bearing colon tumors, as well as rabbits bearing widespread metastases from VX2 systemic arterial disseminated metastases. All the conjugates showed significant T1 and T2 relaxivities. However, the conjugate containing 3-Gd(III)-aminoethylamido-DOTA at position 17 of HPPH demonstrated great potential for tumor imaging by both MR and fluorescence while maintaining its PDT efficacy. At an MR imaging dose (10 µmol/kg), HPPH-3Gd(III)DOTA did not cause any significant organ toxicity in mice, indicating its potential as a cancer imaging (MR and fluorescence) agent with an option to treat cancer by photodynamic therapy (PDT).


Subject(s)
Colonic Neoplasms , Photochemotherapy , Animals , Chlorophyll/analogs & derivatives , Chlorophyll/pharmacology , Chlorophyll A , Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/drug therapy , Heterocyclic Compounds, 1-Ring , Humans , Mice , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Rabbits , Rats
11.
Article in English | MEDLINE | ID: mdl-36568335

ABSTRACT

To investigate the impact of mono- and di-ß-galactose moieties in tumor uptake and photodynamic therapy (PDT) efficacy, HPPH [3-(1'-hexyloxy)ethyl-3-devinylpyropheophorobide-a], the meso pyropheophorbide-a [3-ethyl-3-devinyl-pyropheophorbide-a], and the corresponding 20-benzoic acid analogs were used as starting materials. Reaction of the intermediates containing one or two carboxylic acid functionalities with 1-aminogalactose afforded the desired 172- or 20(4')- mono- and 172, 20(4')-di galactose conjugated photosensitizers (PSs) with and without a carboxylic acid group. The overall lipophilicity caused by the presence of galactose in combination with either an ethyl or (1'-hexyloxy)ethyl side chain at position-3 of the macrocycle made a significant difference in in vitro uptake by tumor cells and photoreaction upon light exposure. Interestingly, among the PSs investigated, compared to HPPH 1 the carbohydrate conjugates 2 and 11 in which ß-galactose moieties are conjugated at positions 172 and 20(4') of meso-pyro pheophorbide-a showed similar in vitro efficacy in FaDu cell lines, but in SCID mice bearing FaDu tumors (head & neck) Ps 11 gave significantly improved long-term tumor cure.

12.
J Med Chem ; 64(1): 741-767, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33400524

ABSTRACT

Erlotinib was covalently linked to 3-(1'-hexyloxy)ethyl-3-devinylpyropheophorbide-a (HPPH) and structurally related chlorins and bacteriochlorins at different positions of the tetrapyrrole ring. The functional consequence of each modification was determined by quantifying the uptake and subcellular deposition of the erlotinib conjugates, cellular response to therapeutic light treatment in tissue cultures, and in eliminating of corresponding tumors grown as a xenograft in SCID mice. The experimental human cancer models the established cell lines UMUC3 (bladder), FaDu (hypopharynx), and primary cultures of head and neck tumor cells. The effectiveness of the compounds was compared to that of HPPH. Furthermore, specific functional contribution of the carboxylic acid side group at position 172 and the chiral methyl group at 3(1') to the overall activity of the chimeric compounds was assessed. Among the conjugates investigated, the PS 10 was identified as the most effective candidate for achieving tumor cell-specific accumulation and yielding improved long-term tumor control.


Subject(s)
Erlotinib Hydrochloride/chemistry , Photosensitizing Agents/chemical synthesis , Porphyrins/chemistry , Animals , Cell Line, Tumor , Cell Survival/drug effects , Coculture Techniques , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/pathology , Humans , Mice , Mice, SCID , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Stereoisomerism , Structure-Activity Relationship , Survival Rate
13.
Chemotherapy ; 56(3): 223-33, 2010.
Article in English | MEDLINE | ID: mdl-20551639

ABSTRACT

BACKGROUND: The purpose of this study was: (1) to document the critical requirement of cystine for growth of human tumor cells in vitro, and (2) to determine the effect of the anticancer agent irinotecan on the cystine transporter x(c)(-) in head and neck FaDu xenografts. METHODS: Cell growth was measured by sulforhodamine B assay. xCT protein, glutathione (GSH) and DNA damage were determined using Western blot, spectrophotometry, and immunohistochemistry, respectively. RESULTS: Depletion of cystine from the medium inhibited tumor cell growth. Treatment of FaDu tumor with a therapeutic dose of irinotecan resulted in depression of xCT protein levels, leading to tumor growth retardation and downregulation of GSH with increased reactive oxygen species (ROS). The accumulation of ROS correlated with increased DNA damage as evidenced by increased H2AX. CONCLUSION: Depression of xCT protein by irinotecan resulted in downregulation of GSH and increase in ROS, which could be the other possible mechanisms of DNA damage by irinotecan.


Subject(s)
Amino Acid Transport Systems/metabolism , Camptothecin/analogs & derivatives , Cystine/physiology , Down-Regulation/physiology , Head and Neck Neoplasms/metabolism , Xenograft Model Antitumor Assays , Amino Acid Transport Systems/antagonists & inhibitors , Amino Acid Transport Systems/biosynthesis , Animals , Camptothecin/pharmacology , Cell Line, Tumor , Cystine/antagonists & inhibitors , Cystine/metabolism , Down-Regulation/drug effects , Female , Humans , Irinotecan , Mice , Mice, Nude , Xenograft Model Antitumor Assays/methods
14.
Photochem Photobiol ; 96(3): 625-635, 2020 05.
Article in English | MEDLINE | ID: mdl-31738460

ABSTRACT

To determine the impact of delivery vehicles in photosensitizing efficacy of HPPH, a hydrophobic photosensitizer was dissolved in various formulations: 1% Tween 80/5% dextrose, Pluronic P-123 and Pluronic F-127 in 0.5%, 1% and 2% phosphate buffer solutions (PBS). HPPH was also conjugated to Pluronic F-127, and the resulting conjugate (PL-20) was formulated in PBS. Among the different delivery vehicles, only Pluronic P-123 displayed significant vehicle cytotoxicity, whereas Pluronic F127 was nontoxic. Compared to PL-20, HPPH formulated in Tween80 and Pluronic F-127 showed higher cell-uptake, but lower long-term retention in Colon26 cell compared to PL-20. The higher retention of PL-20 was similarly observed during in vivo uptake with BALB/c mice baring Ct26 tumors. In contrast to the in vitro uptake experiments, PL-20 showed slightly higher uptake compared to HPPH formulated in Tween or Pluronic-F127. A significant difference in pharmacokinetic profile was also observed between the HPPH-Pluronic formulation and PL-20. Under similar in vivo treatment parameters (drug dose 0.47 µmol kg-1 , light dose: 135 J cm-2 at 24 h post-injection of PS), HPPH formulated either in Tween or Pluronic F-127 formulation showed similar in vivo PDT efficacy (20-30% tumor cure on day 60), whereas PL-20 showed 40% tumor cure (day 60).


Subject(s)
Chlorophyll/analogs & derivatives , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Poloxamer/administration & dosage , Animals , Cell Line, Tumor , Chlorophyll/administration & dosage , Humans , Mice , Mice, Inbred BALB C , Xenograft Model Antitumor Assays
15.
Sci Rep ; 10(1): 21791, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33311561

ABSTRACT

This article presents the construction of a multimodality platform that can be used for efficient destruction of brain tumor by a combination of photodynamic and sonodynamic therapy. For in vivo studies, U87 patient-derived xenograft tumors were implanted subcutaneously in SCID mice. For the first time, it has been shown that the cell-death mechanism by both treatment modalities follows two different pathways. For example, exposing the U87 cells after 24 h incubation with HPPH [3-(1'-hexyloxy)ethyl-3-devinyl-pyropheophorbide-a) by ultrasound participate in an electron-transfer process with the surrounding biological substrates to form radicals and radical ions (Type I reaction); whereas in photodynamic therapy, the tumor destruction is mainly caused by highly reactive singlet oxygen (Type II reaction). The combination of photodynamic therapy and sonodynamic therapy both in vitro and in vivo have shown an improved cell kill/tumor response, that could be attributed to an additive and/or synergetic effect(s). Our results also indicate that the delivery of the HPPH to tumors can further be enhanced by using cationic polyacrylamide nanoparticles as a delivery vehicle. Exposing the nano-formulation with ultrasound also triggered the release of photosensitizer. The combination of photodynamic therapy and sonodynamic therapy strongly affects tumor vasculature as determined by dynamic contrast enhanced imaging using HSA-Gd(III)DTPA.


Subject(s)
Brain Neoplasms/therapy , Chlorophyll/analogs & derivatives , Photochemotherapy , Ultrasonic Waves , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Chlorophyll/pharmacology , Mice , Mice, SCID , Xenograft Model Antitumor Assays
16.
J Photochem Photobiol B ; 211: 111998, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32862090

ABSTRACT

Herein we report the positron emission tomography (PET) imaging potential of a 124I-labeled radiopharmaceutical (PET-ONCO). In tumored mice, it shows high uptake in a variety of tumors: brain (GL261, U87), Colon (Colon26), lung (Lewis lung), breast (4 T1), bladder (UMUC3), pancreas (PANC-1) implanted in mice. This agent also shows promise for imaging associated metastatic disease (breast to lung, to bone). Interestingly, the iodinated compound derived from chlorophyll-a, in combination with the corresponding 124I-analog, can serve as a dual imaging agent (PET/fluorescence, complimentary to each other), with an option of photodynamic therapy (PDT). In contrast to Fluorine-18 (half-life 110 min), the Iodine-124 radionuclide has a physical half-life of roughly 4 days. Thus, unlike 18F-FDG, PET-ONCO can be transported longer distances. While the time for optimal tumor-uptake was observed at 24 h, improved tumor contrasts of both primary and metastasis were obtained at 48 and 72 h post- injection (i. v.) of PET-ONCO. In both mice and rats at a single dose study, PET-ONCO did not show any organ toxicity.


Subject(s)
Chlorophyll A/chemistry , Indicators and Reagents/chemistry , Neoplasms/diagnostic imaging , Neoplasms/radiotherapy , Animals , Biological Transport , Chlorophyll A/metabolism , Female , Fluorine Radioisotopes/chemistry , Humans , Iodine Radioisotopes/chemistry , Male , Mice, Inbred BALB C , Optical Imaging , Photochemotherapy , Porphyrins/chemistry , Positron-Emission Tomography , Rats, Sprague-Dawley , Time Factors
17.
J Med Chem ; 62(5): 2598-2617, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30776232

ABSTRACT

The in vitro and in vivo anticancer activity of iodinated photosensitizers (PSs) with and without an erlotinib moiety was investigated in UMUC3 [epidermal growth factor (EGFR)-positive] and T24 (EGFR-low) cell lines and tumored mice. Both the erlotinib-conjugated PSs 3 and 5 showed EGFR target specificity, but the position-3 erlotinib-PS conjugate 3 demonstrated lower photodynamic therapy efficacy than the corresponding non-erlotinib analogue 1, whereas the conjugate 5 containing an erlotinib moiety at position-17 of the PS showed higher tumor uptake and long-term tumor cure (severe combined immunodeficient mice bearing UMUC3 tumors). PS-erlotinib conjugates in the absence of light were ineffective in vitro and in vivo, but robust apoptotic and necrotic cell death was observed in bladder cancer cells after exposing them to a laser light at 665 nm. In contrast to 18F-fluorodeoxyglucose, a positron emission tomography agent, the position-17 erlotinib conjugate (124I-analogue 6) showed enhanced UMUC3 tumor contrast even at a low imaging dose of 15 µCi/mouse.


Subject(s)
Photochemotherapy , Photosensitizing Agents/pharmacology , Urinary Bladder Neoplasms/diagnostic imaging , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , ErbB Receptors/drug effects , Erlotinib Hydrochloride/pharmacology , Erlotinib Hydrochloride/therapeutic use , Humans , Mice , Mice, SCID , Photosensitizing Agents/therapeutic use , Positron-Emission Tomography , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
18.
Clin Cancer Res ; 13(3): 965-71, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17289892

ABSTRACT

PURPOSE: Chemotherapy-induced diarrhea occurs secondary to mucosal inflammation and may be cyclooxygenase-2 mediated. Cyclooxygenase-2 inhibitors may ameliorate chemotherapy-induced mucosal toxicity and enhance its antitumor effect. We investigated this hypothesis in the Ward colorectal cancer rat model and in a phase I clinical study. EXPERIMENTAL DESIGN: In the Ward rat model, irinotecan was given daily x 3 or weekly x 4 with or without celecoxib. In the phase I clinical study, we planned to escalate the dose of irinotecan in the FOLFIRI regimen (irinotecan, 5-fluorouracil, and leucovorin) with a fixed dose of celecoxib. Irinotecan was escalated in four dose levels: 180, 200, 220, and 260 mg/m2. Celecoxib was administered as 400 mg, twice daily starting on day 2 of cycle 1. Pharmacokinetics of irinotecan, SN-38, and SN-38G were obtained on days 1 and 14. A standard 3+3 dose escalation scheme was used. Plasma concentrations of irinotecan, SN-38, and SN-38G were measured using high-pressure liquid chromatography. RESULTS: Celecoxib ameliorated diarrhea, weight loss, and lethality and resulted in synergistic antitumor effect in the rat model. Twelve patients with advanced cancers were enrolled and evaluable for dose-limiting toxicity (DLT). Diarrhea was the cause for discontinuation in one. Grade 2 and 3 diarrhea occurred in three and two patients, respectively. One patient had DLT at dose level 2 (grade 3 diarrhea). Two had a DLT at DL3 (G3 emesis and myocardial infarct). Celecoxib had limited influence on the pharmacokinetics of irinotecan in this data set. CONCLUSIONS: Maximum tolerated dose of irinotecan in FOLFIRI schedule with celecoxib is 200 mg/m2.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Colorectal Neoplasms/drug therapy , Fluorouracil/administration & dosage , Intestinal Mucosa/drug effects , Mucositis/chemically induced , Neoplasms, Experimental/drug therapy , Pyrazoles/administration & dosage , Pyrazoles/therapeutic use , Sulfonamides/administration & dosage , Sulfonamides/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Camptothecin/administration & dosage , Camptothecin/therapeutic use , Celecoxib , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Fluorouracil/therapeutic use , Glucuronates/administration & dosage , Humans , Intestinal Mucosa/pathology , Irinotecan , Leucovorin/therapeutic use , Male , Maximum Tolerated Dose , Neoplasm Transplantation , Rats , Rats, Inbred F344
19.
Biochem Pharmacol ; 73(9): 1280-7, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17239826

ABSTRACT

This study was designed to understand the basis for the efficacy of methylselenocysteine (MSC) in increasing the therapeutic index of irinotecan against human tumor xenografts. Nude mice bearing human head and neck squamous cells carcinoma xenografts (FaDu and A253) were treated orally with different doses of MSC and irinotecan. Plasma, tumor and normal tissue samples were collected at different times after MSC treatments and were analyzed for selenium (Se) concentration using electrothermal atomic absorption spectrophotometry. MSC is highly effective in modulating the therapeutic index of irinotecan. Enhanced irinotecan efficacy was greater in FaDu tumors (100% CR) than in A253 tumors (60% CR), and depended on MSC dose with a minimum effective dose of 0.01 mg/dx28. The highest plasma Se concentration was achieved 1h after a single dose and 28 d after daily treatments of MSC. The ability of FaDu tumors to retain Se was significantly better than A253 tumors, and the highest Se concentration in normal tissue was achieved in the liver. Peak plasma and tissue Se concentrations were functions of the dose and duration of MSC treatment. The MSC-dependent increase in Se level in normal tissues may contribute to the protective effect against irinotecan toxicity observed in those tissues. Intratumoral total Se concentration was not found to be predictive of the combination therapy response rates. There is a critical need to develop a method to measure the active metabolite of MSC, rather than total Se.


Subject(s)
Camptothecin/analogs & derivatives , Cysteine/analogs & derivatives , Neoplasms/drug therapy , Organoselenium Compounds/pharmacology , Plasma/drug effects , Selenium/blood , Administration, Oral , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cysteine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Female , Humans , Irinotecan , Kidney/drug effects , Kidney/metabolism , Kinetics , Liver/drug effects , Liver/metabolism , Mice , Mice, Nude , Plasma/chemistry , Plasma/metabolism , Selenium/metabolism , Selenium/pharmacokinetics , Selenocysteine/analogs & derivatives
20.
Mol Cancer Ther ; 4(5): 843-54, 2005 May.
Article in English | MEDLINE | ID: mdl-15897249

ABSTRACT

The combination of methylselenocysteine and irinotecan (CPT-11) is synergistic against FaDu and A253 xenografts. Methylselenocysteine/CPT-11 increased tumor cure rate to 100% in FaDu and to 60% in A253. In this study, the effect of methylselenocysteine on pharmacokinetic and pharmacogenetic profiles of genes relevant to CPT-11 metabolic pathway was evaluated to identify possible mechanisms associated with the observed combinational synergy. Nude mice bearing tumors (FaDu and A253) were treated with methylselenocysteine, CPT-11, and a combination of methylselenocysteine/CPT-11. Samples were collected and analyzed for plasma and intratumor concentration of CPT-11 and 7-ethyl-10-hydroxyl-camptothecin (SN-38) by high-performance liquid chromatography. The intratumor relative expression of genes related to the CPT-11 metabolic pathway was measured by real-time PCR. After methylselenocysteine treatment, the intratumor area under the concentration-time curve of SN-38 increased to a significantly higher level in A253 than in FaDu and was associated with increased expression of CES1 in both tumors. Methylselenocysteine/CPT-11 treatment, compared with CPT-11 alone, resulted in a significant decrease in levels of ABCC1 and DRG1 in FaDu tumors and an increase in levels of CYP3A5 and TNFSF6 in A253 tumors. No statistically significant changes induced by methylselenocysteine/CPT-11 were observed in the levels of other investigated variables. In conclusion, the significant increase in the cure rate after methylselenocysteine/CPT-11 could be related to increased drug delivery into both tumors (CES1), reduced resistance to SN-38 (ABCC1 and DRG1) in FaDu, and induced Fas ligand apoptosis (TNFSF6) in A253. No correlation was observed between cure rate and other investigated variables (transporters, degradation enzymes, DNA repair, and cell survival/death genes) in either tumor.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Biomarkers, Tumor/metabolism , Camptothecin/analogs & derivatives , Carcinoma, Squamous Cell/drug therapy , Cysteine/analogs & derivatives , Head and Neck Neoplasms/drug therapy , Pharmacogenetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Camptothecin/administration & dosage , Carcinoma, Squamous Cell/blood , Carcinoma, Squamous Cell/genetics , Cysteine/administration & dosage , Female , Head and Neck Neoplasms/blood , Head and Neck Neoplasms/genetics , Humans , Irinotecan , Mice , Mice, Nude , Organoselenium Compounds/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Selenocysteine/analogs & derivatives , Transplantation, Heterologous , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL