18 results on '"Izabela Tworowska"'
Search Results
2. 64Cu-DOTATATE PET/CT for Imaging Patients with Known or Suspected Somatostatin Receptor–Positive Neuroendocrine Tumors: Results of the First U.S. Prospective, Reader-Masked Clinical Trial
- Author
-
Ayman M. Gaber, Rodolfo Nunez, Andreas Kjaer, Afshin Shafie, David Ranganathan, Ali Abbasi, Nilesh K. Wagh, Ebrahim S. Delpassand, and Izabela Tworowska
- Subjects
PET-CT ,Somatostatin receptor ,business.industry ,Neuroendocrine tumors ,medicine.disease ,030218 nuclear medicine & medical imaging ,Clinical trial ,03 medical and health sciences ,0302 clinical medicine ,64Cu-DOTATATE ,030220 oncology & carcinogenesis ,Medicine ,Radiology, Nuclear Medicine and imaging ,Nuclear medicine ,business ,Prospective cohort study ,Adverse effect ,Somatostatin Receptor Positive - Abstract
Studies demonstrate that the investigational 64Cu-DOTATATE radiopharmaceutical may provide diagnostic and logistical benefits over available imaging agents for patients with somatostatin receptor (SSTR)–positive neuroendocrine tumors (NETs). Accordingly, we aimed to prospectively determine the lowest dose of 64Cu-DOTATATE that facilitates diagnostic-quality scans and evaluated the diagnostic performance and safety in a phase III study of patients with SSTR-expressing NETs. Methods: A dose-ranging study was conducted on 12 patients divided into 3 dose groups (111 MBq [3.0 mCi], 148 MBq [4.0 mCi], and 185 MBq [5.0 mCi] ± 10%) to determine the lowest dose of 64Cu-DOTATATE that produced diagnostic-quality PET/CT images. Using the 64Cu-DOTATATE dose identified in the dose-ranging study, 3 independent nuclear medicine physicians who were masked to all clinical information read PET/CT scans from 21 healthy volunteers and 42 NET-positive patients to determine those with disease or no disease, as well as those with localized versus metastatic status. Masked-reader evaluations were compared with a patient-specific standard of truth, which was established by an independent oncologist who used all previously available pathology, clinical, and conventional imaging data. Diagnostic performance calculated for 64Cu-DOTATATE included sensitivity, specificity, negative predictive value, positive predictive value, and accuracy. Inter- and intrareader reliability, as well as ability to differentiate between localized and metastatic disease, was also determined. Adverse events were recorded from 64Cu-DOTATATE injection through 48 h after injection. Results: The dose-ranging study identified 148 MBq (4.0 mCi) as the optimal dose to obtain diagnostic-quality PET/CT images. After database lock, diagnostic performance from an initial majority read of the 3 independent readers showed a significant 90.9% sensitivity (P = 0.0042) and 96.6% specificity (P
- Published
- 2020
- Full Text
- View/download PDF
3. Preclinical Investigation of 212Pb-DOTAMTATE for Peptide Receptor Radionuclide Therapy in a Neuroendocrine Tumor Model
- Author
-
Izabela Tworowska, Ebrahim S. Delpassand, Amal Saidi, Tania Stallons, and Julien Torgue
- Subjects
0301 basic medicine ,Cancer Research ,Receptors, Peptide ,Octreotide ,Pharmacology ,Neuroendocrine tumors ,Article ,Heterocyclic Compounds, 1-Ring ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Acetamides ,medicine ,Animals ,Humans ,Receptor ,Radioisotopes ,Octreotate ,Somatostatin receptor ,business.industry ,Ascorbic acid ,medicine.disease ,Neuroendocrine Tumors ,030104 developmental biology ,Somatostatin ,Lead ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Radionuclide therapy ,Fluorouracil ,Radiopharmaceuticals ,business ,medicine.drug - Abstract
Somatostatin analogues have been examined as a treatment for somatostatin receptor overexpressing tumors for years; specifically, octreotate (TATE) and octreotide (TOC). Several versions of these analogues coupled to beta or gamma nuclides are currently used as imaging agents, as treatments with peptide receptor radionuclide therapy (PRRT) for patients with neuroendocrine tumors or are being explored in preclinical and clinical settings. Our study describes the use of 212Pb-DOTAMTATE, the octreotate analogue, in combination with 212Pb, the parent of an alpha emitter. Preclinical studies demonstrated tumor targeting of 212Pb-DOTAMTATE of >20% ID/g up to 24 hours post drug injection. The addition of kidney protection agents, including l-lysine and l-arginine decreases drug accumulation in the kidneys and the addition of ascorbic acid to the chelation mixture reduces oxidation of the drug product. 212Pb-DOTAMTATE displays a favorable toxicity profile with single-dose injections of 20 μCi showing 100% survival and with nontoxic cumulative doses up to 45 μCi, when fractionated into three smaller doses of 15 μCi. In an initial efficacy study, a single 10 μCi injection of 212Pb-DOTAMTATE extended the mean survival 2.4-fold. Efficacy was enhanced by giving three treatment cycles of 212Pb-DOTAMTATE and reducing the time between injections to two weeks. Efficacy was optimized further by the addition of a chemo-sensitizing agent, 5-fluorouracil, given in combination with three cycles of 10 μCi 212Pb-DOTAMTATE. These conditions led to 79% of the animals being tumor free at the end of the 31-week study suggesting that 212Pb-DOTAMTATE alone or in combination with a chemotherapeutic may have positive clinical implications.
- Published
- 2019
- Full Text
- View/download PDF
4. Phase I dose-escalation study of AlphaMedix for targeted-alpha-emitter therapy of PRRT-naive neuroendocrine patients
- Author
-
Jason Daniel Hurt, Julien Torgue, Ebrahim S. Delpassand, Rouzbeh Esfandiari, Izabela Tworowska, and Rodolfo Nunez
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Internal medicine ,Dose escalation ,Medicine ,Alpha (ethology) ,business ,Objective response ,Common emitter - Abstract
4117 Background: Peptide-receptor-radioligand-therapy (PRRT) has been shown to increase the progression-free-survival (PFS) of neuroendocrine patients, however the objective response rate is rather low. The targeted-alpha-emitter therapy (TAT) of NETs can increase the ORR and induce partial or complete tumor response. In this abstract, we present results of the safety and efficacy of AlphaMedix (212Pb-DOTAMTATE) done in PRRT-naïve patients with SSTR-expressing NETs (FDA IND 135150). Methods: The phase 1 dose escalation study (IND 135150) was designed according to a 3+3 dose-escalation scheme (30% increase of the dose in each subsequent cohort). We enrolled PRRT naïve subjects with biopsy-proven unresectable or metastatic SSTR-expressing NETs from different primary sites (midgut, rectal, pancreas, and lung) with at least one measurable lesion. Response to treatment was measured per RECIST 1.1 criteria and the effect on quality of life was measured with the EORTC-QLQ-C30 QOL questionnaire. Results: A total of 20 PRRT naïve subjects (10 men and 10 women; median age 60 ( range 27-80)) have been treated to date. 10 subjects in MAD4 cohort received at least three cycles of 212Pb-DOTAMTATE at the highest dose level of 67.6 µCi/kg/cycle. Of these, 6/10 subjects (60%) have completed all four-cycles of treatment. Radiographic evaluation of these six-MAD4 patients reveal an ORR in 5 out of 6 subjects (83.3%) per RECIST 1.1 criteria (1CR, 4PR, 1SD) in addition to a 100% response according to 68Ga-DOTATATE-PET/CT-imaging, defined as complete resolution of SSTR-positive lesions (CR) or Partial Response (PR) defined as resolution of most active lesions or substantially decreased SUV (>25%). No progression of disease was noted in the first six subjects enrolled in the MAD4 cohort who have completed treatment. All six-patients have 100% PFS up to 22 months (for the first 3-subjects in MAD4) and up to 19 months (next 3-patients). Except for mild-to-moderate, reversible hair loss, there were no other clinically significant drug related AE, or SAE. The most frequent AEs of any grade (> 4 subjects) reported include fatigue (35%), hyperglycemia (30%), lymphopenia (30%) alopecia (30%), and diarrhea (20%). Five grade 3 AEs were reported (back pain, dysarthria, dyspnea, acute kidney injury), no grade 4 AEs, and one grade 5 AE was reported. There was a significant improvement in the quality of life, reduction of pain, shortness of breath in the majority of subjects, with increase of energy. Conclusions: This F-I-H clinical study of AlphaMedix shows that PRRT with 212Pb is feasible, well tolerated, and provides substantial reduction in tumor burden to patients with unresectable, metastatic SSTR-expressing NETs. Dramatic improvement in tumor burden and a positive impact on quality of life were seen in all of the PRRT naïve subjects who AlphaMedix at the highest dose tested. Clinical trial information: NCT03466216.
- Published
- 2021
- Full Text
- View/download PDF
5. Abstract CT159: First-in-human dose escalation of AlphaMedixTM for targeted alpha-emitter therapy of neuroendocrine tumors
- Author
-
Farah Shanoon, Julien Torgue, Ebrahim S. Delpassand, Rodolfo Nunez, Izabela Tworowska, and J.D. Hurt
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,Nausea ,Cancer ,Neuroendocrine tumors ,medicine.disease ,Gastroenterology ,Oncology ,Quality of life ,Internal medicine ,Cohort ,Toxicity ,medicine ,Dosing ,medicine.symptom ,business ,Adverse effect - Abstract
Introduction: Peptide Receptor Radioligand Therapy (PRRT) has been shown to be an effective treatment for patients with metastatic somatostatin receptor (SSTR) positive neuroendocrine tumors (NETs), however interest in developing alpha-emitter based therapies remains high. We present the initial safety and preliminary efficacy of this first-in-human (FIH) study of AlphaMedix™ (212Pb-DOTAMTATE), a novel somatostatin analog for Targeted Alpha-emitter Therapy (TAT), in patients with SSTR expressing NETs (FDA IND 135150). Methods: Thirteen adult subjects, 6 men and 7 women, median age 68 (range 27-75), with biopsy-proven unresectable or metastatic SSTR (+) NETs from different primary sites (small bowel, pancreas, and lung) with at least one measurable lesion were treated with a Single, weight-based, Ascending Dose (SAD) of AlphaMedix™. Dose escalation was conducted according to a classic 3+3 design. Once a partial response was observed in the SAD-3 cohort, the Multiple Ascending Dosing (MAD) began at the same dose level (3 cycles dosed every 8-weeks). Subjects who had previously received PRRT were excluded. All patients received amino acid renal protection prior to AlphaMedixTM administration. Response to treatment was measured per RECIST 1.1 and the effect on the quality of life was measured with the EORTC-QLQ-C30 QOL questionnaire. Two SAD cohorts (SAD1 and SAD2) received 30.7 and 40.0 µCi/kg respectively. MAD3 received 52.0 µCi/kg per cycle and MAD4, began dosing at 67.6 µCi/ kg for a total dose ranging from 14.7 to 16.8 mCi, across 3 cycles. Results: All 3 subjects in MAD 4 who received 67.6 µCi/ kg for 3 cycles showed partial response with 73%, 71%, and 33% decrease in size of the index lesions respectively. 68Ga DOTATATE PET/CT revealed almost a complete response in 2 subjects and a partial response in the third. No clinically significant investigational drug-related hematological and renal toxicity was noted. The most common adverse events noted were diarrhea 2/13(23%), nausea 4/13(30%), fatigue 4/13(30%), hyperglycemia 7/13(53%). Moderate hair loss was seen in 2/13 (15%) patients. Quality of life parameters suggest significant improvement in pain, energy, and shortness of breath in the majority of subjects. Conclusion: Dramatic decreases in tumor burden and a positive impact on quality of life were seen in all of the subjects who received three cycles of AlphaMedixTM at the highest dose tested. In addition, AlphaMedixTM was extremely well tolerated with only mild adverse events, most of which were attributable to the AA solution used for renal protection. This FIH study of AlphaMedixTM illustrates that PRRT with 212Pb is feasible, well-tolerated, and provides substantial reduction in tumor burden to patients with unresectable, metastatic SSTR expressing NETs Citation Format: Izabela Tworowska, Ebrahim S. Delpassand, Julien Torgue, Farah Shanoon, Jason Hurt, Rodolfo Nunez. First-in-human dose escalation of AlphaMedixTM for targeted alpha-emitter therapy of neuroendocrine tumors [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr CT159.
- Published
- 2020
- Full Text
- View/download PDF
6. Targeted Alpha-emitter Therapy of Neuroendocrine Tumors using 212Pb-octreotate (AlphaMedix TM)
- Author
-
Farah Shanoon, Tania Stallons, Ebrahim S. Delpassand, Ali Muzammil, Luke Bolek, Michael Ghaly, Bin He, Eric C. Frey, Amal Saidi, Izabela Tworowska, Julien Torgue, and George Sgouros
- Subjects
Octreotate ,chemistry.chemical_compound ,chemistry ,Radiological and Ultrasound Technology ,business.industry ,Cancer research ,medicine ,Alpha (ethology) ,Radiology, Nuclear Medicine and imaging ,Neuroendocrine tumors ,medicine.disease ,business ,Common emitter - Published
- 2019
- Full Text
- View/download PDF
7. Theranostic 203/212Pb-labeled octreotate analogs (AlphaMedix TM) and their preclinical characterization for the Phase I clinical studies in neuroendocrine cancer patients
- Author
-
Julien Torgue, Amal Saidi, Garry E. Kiefer, Izabela Tworowska, Tania Stallons, George Sgouros, Ebrahim S. Delpassand, F. Rojas-Quijano, Mickel Ghaly, Eric C. Frey, and Paul Jurek
- Subjects
Cancer Research ,Octreotate ,chemistry.chemical_compound ,chemistry ,business.industry ,Neuroendocrine Cancer ,Cancer research ,Molecular Medicine ,Medicine ,Radiology, Nuclear Medicine and imaging ,business - Published
- 2019
- Full Text
- View/download PDF
8. Safety and Effectiveness of 177Lu-DOTATATE Peptide Receptor Radionuclide Therapy After Regional Hepatic Embolization in Patients With Somatostatin-Expressing Neuroendocrine Tumors
- Author
-
Muzammil Ali, Izabela Tworowska, Mohammadali Hamiditabar, Ebrahim S. Delpassand, Luke Bolek, and Gelareh Vahdati
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,Peptide receptor ,Adolescent ,Receptors, Peptide ,medicine.medical_treatment ,Neuroendocrine tumors ,Octreotide ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Internal medicine ,177Lu-DOTATATE ,medicine ,Organometallic Compounds ,Humans ,Radiology, Nuclear Medicine and imaging ,In patient ,Embolization ,Receptor ,Child ,Aged ,Retrospective Studies ,Aged, 80 and over ,business.industry ,General Medicine ,Middle Aged ,medicine.disease ,Embolization, Therapeutic ,Gene Expression Regulation, Neoplastic ,Neuroendocrine Tumors ,Somatostatin ,Liver ,030220 oncology & carcinogenesis ,Radionuclide therapy ,Female ,Radiology ,business - Abstract
Peptide receptor radionuclide therapy (PRRT) with Lu-DOTATATE is shown to be an effective therapeutic option for somatostatin-expressing neuroendocrine neoplasms. Some concerns are raised over safety of this modality in patients with a history of regional chemoembolization and radionuclide hepatic embolization (CRHE) and is cause of reluctance among some physicians for suggesting Lu-DOTATATE in this patient population.We retrospectively reviewed 143 patients with somatostatin-expressing neuroendocrine tumors who underwent Lu-DOTATATE PRRT. Statistical analysis was performed on effect of Lu-DOTATATE in patients with and without prior CRHE using resampling procedures and correlation coefficient (r).Proportion of toxicity in patients with and without CRHE was comparable (P = 0.246). No statistically significant correlation (r) found between any toxicity and prior CRHE (r = -0.3 to -0.03) or time elapsed between embolization and the first cycle of PRRT (r = -0.59 to 0.17). Following PRRT, 76.5% of patients with CRHE experienced benefit (partial response + stable disease), whereas 23.4% experienced progressive disease. Patients with CRHE showed more stable disease (P = 0.048) and less progressive disease (P = 0.046) following PRRT compared with no CRHE. The CRHE and no-CRHE status shared same probability for developing partial response/complete response following PRRT (P = 0.50).Treatment with Lu-DOTATATE did not show clinically or statistically significant toxicity in CRHE patients regardless of frequency of embolization or time interval between embolization and first PRRT. Results suggested a statistically significant higher response rate in patients with a history of CRHE. A prior history of CRHE is not a contraindication to subsequent PRRT.
- Published
- 2017
9. Peptide Receptor Radionuclide Therapy With 177Lu-Octreotate in Patients With Somatostatin Receptor Expressing Neuroendocrine Tumors: Six Years' Assessment
- Author
-
Edward M. Wolin, Thomas M. O'Dorisio, Muzammil Ali, David Ranganathan, Izabela Tworowska, Ebrahim S. Delpassand, Mohammadali Hamiditabar, Joseph Roys, and Jonathan R. Strosberg
- Subjects
Adult ,Male ,medicine.medical_specialty ,Peptide receptor ,Adolescent ,Neuroendocrine tumors ,Octreotide ,Disease-Free Survival ,Article ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,In patient ,Progression-free survival ,Receptors, Somatostatin ,Neoplasm Metastasis ,Receptor ,Child ,Aged ,Aged, 80 and over ,Somatostatin receptor ,business.industry ,General Medicine ,Middle Aged ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Neuroendocrine Tumors ,Endocrinology ,Somatostatin ,030220 oncology & carcinogenesis ,Radionuclide therapy ,Cancer research ,Female ,business - Abstract
Peptide receptor radionuclide therapy (PRRT) with radiolabeled somatostatin analogues is a promising treatment for patients with inoperable, well to moderately differentiated metastatic neuroendocrine tumors (NETs). In continuation of our novel study with the radionuclide lutetium Lu, we now present further results of Lu DOTATATE therapy in managing NETs and other somatostatin receptor-expressing tumors in a larger and more diverse patient group.One hundred forty-four consecutive patients (85 men and 59 women; age range, 11-87 years; mean age, 58.5 years) with histologically confirmed NET were enrolled. One hundred forty-three patients received at least 1 cycle of treatment. Among them, 132 were deemed evaluable by having at least 1 cycle of treatment and a posttreatment MRI or CT scan for assessment based on modified Response Evaluation Criteria in Solid Tumors. Response to therapy was evaluated in terms of progression-free survival, overall survival, as well as radiologic, biochemical, and clinical responses. Further, analysis of symptoms was reviewed during therapy and also in subsequent follow-ups for safety evaluation. Renal, gastrointestinal (GI), hepatic, and hematological adverse events were evaluated using National Cancer Institute common toxicities criteria V4.03, through full blood panels, as well as consultation with patients for any symptoms and/or adverse events.As of July 2016, median progression-free survival was about to be reached. Of 28 patients who have completed Lu DOTATATE therapy (completion of 4 or more cycles of treatment and all designated follow-ups), no patient showed complete response (CR), 8 patients (28.57%) showed partial response (PR), 16 patients (57.14%) showed stable disease (SD), and progressive disease (PD) was observed in 4 patients (14.28%). The objective response rate (CR + PR) of this group was 28.57% (n = 8) with a cumulative disease control (CR + PR + SD) of 85.71% (n = 24).Among 132 evaluable patients, assessment of treatment response using modified Response Evaluation Criteria in Solid Tumors criteria revealed CR in none of the patients, PR in 12 patients (9.09%), SD in 66 patients (50%), whereas PD, which included patients who passed away, was observed in 54 patients (40.90%), yielding an objective response rate of 9.09% (n = 12) and a cumulative disease control rate of 59.09% (n = 78).Symptoms including abdominal pain, diarrhea, flushing, and fatigue improved in over 50% of the patients, whereas weight loss improved in 28.26% of the patients. No grade 3 or grade 4 renal toxicities were found, though eleven grade 3 and five grade 4 hematological as well as three grade 3 hepatotoxicities were reported. Grade 3 hematotoxicity lasted an average of 2.7 months, and grade 4 lasted for only 0.9 months, whereas grade 3 hepatotoxicity lasted an average of 3.1 months.Lu-octreotate peptide receptor radionuclide therapy has shown promising potential as a safe and effective targeted therapy in inoperable, well to moderately differentiated metastatic neuroendocrine cancers. The results of the multicenter randomized clinical trial conducted in United States and Europe are concordant with current study.
- Published
- 2017
10. Abstract LB-A16: Imaging of glioblastoma using LDLR-based targeted delivery system
- Author
-
Leo G. Flores, Jamal Temsamani, Rafal Zielinski, Jonathan A. Nowak, Ebrahim S. Delpassand, Izabela Tworowska, Pascaline Lécorché, Cedric Malicet, and Michel Khrestchatisky
- Subjects
Cancer Research ,Biodistribution ,Chemistry ,Brain tumor ,Cancer ,medicine.disease ,Blood–brain barrier ,In vitro ,medicine.anatomical_structure ,Oncology ,In vivo ,LDL receptor ,Cancer research ,medicine ,Receptor - Abstract
Introduction: Glioblastoma is the most agressive brain tumor with average life expectancy of 12-15 months from diagnosis. One of the factors that limit efficacy of drug, especially in primary brain tumor is permeability of the blood brain barrier (BBB). The low-density lipoprotein receptor (LDLR) expressed at the BBB mediates the transport of endogenous ligands through the BBB, a process referred to as receptor-mediated transcytosis. VECT-HORUS (VH) has identified and chemically optimized a family of peptide-vectors targeting both the human and murine LDLR and able i) to cross the BBB and ii) to target tumors such as glioblastoma that express high levels of the LDLR. The objective of this study was to determine the LDLR targeting properties of 68Ga/177Lu- radiolabeled peptide vectors developed by the VH proprietary platform using a glioblastoma model that expresses the human LDLR (hLDLR) at high levels. Methods: The LDLR targeted DOTA-conjugates (VH-DO31, VH-DO33) and NODAGA conjugate (VH-NO31), (10-30ug, VECT-HORUS SAS, France) were labeled with 68Ga (1.5mCi) eluted from 68Ge/68Ga generator (100mCi, ITG GmBH, Germany) or with 177Lu n.c.a (1mCi, ITG GmBH, Germany). The U87MG cell line has been shown to express high levels of the hLDLR. The LDLR targeting properties of these conjugates were thus determined in vitro in U87MG cellular uptake studies, as well as in vivo in U87MG xenografted mice. The PET/CT images of U87MG xenograft generated in athymic nude mice (10 weeks, n=3) were acquired using G4 PET/Xray camera (Sofie Biosciences; 10min/scan) at 1h, 2h, 3h and 4h post-injection. Results: All 68Ga/177Lu-labeled conjugates were synthesized with radiochemical purity higher than 91 % as determined by radio-HPLC. Radiolytic stability of agents was increased using C18 ethanol purification of the final products. 177Lu-VH-DO33 showed the highest retention of the agent in U87MG cell line at 1h (13.88± 1.6 %ID/mg) and 21h incubation time (8.7± 4 %ID/mg) compared to 177Lu-VH-DO31 (8.28 ±6.2 %ID/mg) The microPET imaging studies showed rapid accumulation and retention of all VH derivatives in tumor as monitored up to 4h post injection. All agents were eliminated through bladder and kidneys. There was no accumulation of agents in the bone marrow. The image based biodistribution studies of 68Ga-VH-DO31, 68Ga-VH-DO33 and 68Ga-VH-NO31 showed that the tumor to muscle ratios (SUV ratio) after 30 min post-injection were 4.12, 5.07 and 3.88, respectively and remained at the same levels up to 3h post-injection. The SUV ratios of tumor to kidneys were as follows: 68Ga-VH-DO31 (0.46), 68Ga-VH-DO33 (0.84) and 68Ga-VH-NO31 (0.46) confirming renal elimination of the agents. Conclusions: VH derivatives showed favorable hLDLR and tumor-targeting properties both in in vitro as well as in vivo in U87MG xenografts. These results can postulate that hLDLR may serve as a target for the development of theranostic probes for the diagnosis and radiotherapy of glioblastoma. Citation Format: Izabela Tworowska, Leo G Flores II, Rafal Zielinski, Jonathan Nowak, Pascaline Lecorche, Cedric Malicet, Michel Khrestchatisky, Jamal Temsamani, Ebrahim Delpassand. Imaging of glioblastoma using LDLR-based targeted delivery system [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr LB-A16. doi:10.1158/1535-7163.TARG-19-LB-A16
- Published
- 2019
- Full Text
- View/download PDF
11. Development and Validation of Methods for Quantitative In Vivo SPECT of Pb-212
- Author
-
Izabela Tworowska, George Sgouros, Ebrahim S. Delpassand, Eric C. Frey, Michael Ghaly, and Bin He
- Subjects
Radiological and Ultrasound Technology ,business.industry ,In vivo ,Medicine ,Radiology, Nuclear Medicine and imaging ,Computational biology ,business - Published
- 2019
- Full Text
- View/download PDF
12. P2-09-05: Molecular Imaging of Hedgehog Induced Epithelial-Stromal Interactions
- Author
-
Jennifer Sims-Mourtada, Izabela Tworowska, Richard A. Larson, Firas Mourtada, David J. Yang, Daniel L. Smith, and W.A. Woodward
- Subjects
Patched ,Cancer Research ,Cyclopamine ,biology ,Receptor expression ,Protein degradation ,medicine.disease ,Hedgehog signaling pathway ,Metastasis ,chemistry.chemical_compound ,Oncology ,chemistry ,Cancer research ,biology.protein ,medicine ,Sonic hedgehog ,Hedgehog - Abstract
Introduction: Epithelial-stromal Hedghehog (HH) pathway signaling, originating from Sonic Hedgehog (SHH) ligand secreted by tumor cells, promotes breast cancer growth and metastasis. Furthermore, high tumor SHH expression is associated with poor prognosis in invasive ductal carcinoma. Studies have also shown high expression of Patched (PTCH) mRNA in SHH expressing tumors as well as cancer stem-like cells. This is in part due to a feedback loop that results in increased PTCH gene expression upon HH signaling. We sought to exploit this feedback loop by using radiolabeled derivative of SHH to identify tumors that have a high level of hedgehog signaling. These agents can potentially be used for molecular imaging of HH induced changes in PTCH expression originating from tumor expressed SHH. Methods: Recombinant N-terminal SHH derivatives were radioiodinated with iodine-131 using the idogen method. Iodinated proteins were obtained with radiolabeling yields of 50–60 % and specific activity of 9.3E10Ci/mol. Receptor binding studies were performed on breast cancer cell lines with varying SHH expression. Studies were also conducted on cells cultured in stem cell promoting conditions (mammospheres), after exposure to ionizing radiation or treatment with the hedgehog inhibitor cyclopamine. Scatchard plots were generated using Graph pad software. Pilot studies were conducted with 131I-SHH to test the potential of imaging PTCH receptor expression in a rat model of breast cancer. Fisher rats bearing breast cancer xenografts were injected with approximately 250 microCi of 131I-SHH. Planar scinitigraphy was conducted at 2, 4 and 24 hours. Studies were performed with iodinated BSA as a control for extracellular perfusion and retention. Tissue biodistribution studies were also performed using this model. Results: Receptor binding of 131I-SHH was significantly increased in cell lines with high endogenous HH pathway activation. Receptor binding was also increased after exposure to ionizing radiation and significantly decreased following treatment with hedgehog inhibitors. Scatchard analysis revealed up to a 14 fold increase in PTCH receptor sites in mammospheres compared to the entire breast cancer population. A 10-fold increase in PTCH receptor binding sites was also observed in cells after exposure to ionizing radiation. In vivo imaging and biodistribution studies revealed significant accumulation of 131I-SHH within tumor tissue as compared to normal organs. Tumor:muscle ratios were approximately 8:1 at 4 hours, while tumor to blood and tumor to bone were 2:1 and 5:1 respectively. Significant uptake was also observed in liver tissue, as a result of protein degradation and excretion and endogenous PTCH expression. Conclusions: These studies show that expression of the PTCH receptor is increased in cells with active HH signaling and in breast cancer mammospheres. Our findings also show that PTCH receptor expression is decreased upon treatment with HH signaling inhibitors. Preliminary imaging studies show that 131I-SHH is capable of in vivo detection of breast tumors with high hedgehog signaling. Our data suggests that radiolabeled SHH derivatives may provide a method to follow epithelial-stromal HH interactions and determine response to SHH targeted therapies. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr P2-09-05.
- Published
- 2011
- Full Text
- View/download PDF
13. Abstract 1927: Targeting IL-13RA2 in melanoma and pancreatic cancer
- Author
-
Rafal Zielinski, Waldemar Priebe, Stanislaw Skora, Yue Huang, Izabela Fokt, Waldemar Debinski, Radjendirane Venugopal, Izabela Tworowska, Denise Herpai, Ya'an Kang, Jason B. Fleming, Aleksandra Rusin, and Arumugam Jayakumar
- Subjects
Cancer Research ,business.industry ,Receptor expression ,Melanoma ,Cancer ,medicine.disease ,Interleukin-13 receptor ,Oncology ,In vivo ,Pancreatic cancer ,Cancer research ,Medicine ,Interleukin receptor ,business ,Receptor - Abstract
Background. Interleukin receptor alpha 1 (IL-13RA1) is a component of a heterodimeric IL-13 receptor that is shared with IL-4 and it is ubiquitously expressed in normal tissues and also tumors. In contrast, IL-13RA2 is a monomeric receptor which is overexpressed in various solid tumors, but is virtually absent in normal tissues. Ligands specifically targeting IL-13RA2 but not IL-13RA1, such as Pep-1L and IL-13.E13K.D2.Cys, have been developed and validated [1,2]. Objective. To assess the expression of IL-13RA2 in pancreatic ductal adenocarcinoma and metastatic melanoma specimens and to develop tracers for selective recognition of the tumors. Methods. The expression of IL-13RA2in patient specimens, patient-derived xenografts (PDX) and established cell lines was assessed using Western blot, immunochemistry or immunofluorescence. The ligands were conjugated to a chelator and labeled with Yttrium 86. Ligand specificity was confirmed using IL-13RA2 positive and negative cells and blocking with a “cold” ligand. Biodistribution studies and PET/CT imaging were conducted to assess distribution of the tracer in IL-13RA2 positive tumors models. Results. IL-13RA2 receptor is overexpressed in melanoma and pancreatic cell lines and tumor tissues. Western blot analysis of 56 patients tumor tissue lysates obtained from pancreatic cancer PDX models showed the presence of IL-13RA2 in over 60% of patients. Similarly, tissue microarray analysis of metastatic melanoma samples identified IL-13RA2 receptor expression in 40% of tumor specimens. Uptake study performed in receptor positive and negative cells confirmed the specificity of radiotracer binding. In vivo experiments indicated fast clearance of 86Y- Pep1L ligand with glomerular filtration as a primary mechanism of the tracer removal. Mice bearing IL-13RA2 positive tumors had up taken the tracer with “tumor to muscle” ratio of 5.6 1 h after probe administration. Yttrium 86-labelled IL-13-E13K.D2.Cys was also taken up by IL-13 RA2 positive tumors. Conclusion. IL-13RA2 is an attractive target for the development of comprehensive theranostic strategies for melanoma and pancreatic cancer. References 1. Pandya, H., et al., An interleukin 13 receptor alpha 2-specific peptide homes to human Glioblastoma multiforme xenografts. Neuro Oncol, 2012. 14(1): p. 6-18. 2. Nguyen, V., et al., A novel ligand delivery system to non-invasively visualize and therapeutically exploit the IL13Ralpha2 tumor-restricted biomarker. Neuro Oncol, 2012. 14(10): p. 1239-53. Citation Format: Rafal Zielinski, Izabela Tworowska, Stanislaw Skora, Aleksandra Rusin, Radjendirane Venugopal, Arumugam Jayakumar, Izabela Fokt, Yaan Kang, Jason Fleming, Yue Huang, Denise Herpai, Waldemar Debinski, Waldemar Priebe. Targeting IL-13RA2 in melanoma and pancreatic cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1927.
- Published
- 2018
- Full Text
- View/download PDF
14. Abstract LB-259: Pb203-AR-RMX conjugates for image-guided TAT of neuroendocrine tumors (NETs)
- Author
-
Garry E. Kiefer, Izabela Tworowska, Amal Saidi, Julien Torgue, Paul Jurek, Ebrahim S. Delpassand, Tania Stallons, Federico Rojas-Quijano, and Nilesh K. Wagh
- Subjects
Cancer Research ,Octreotate ,010405 organic chemistry ,business.industry ,Dose fractionation ,Octreotide ,Cancer ,Neuroendocrine tumors ,medicine.disease ,01 natural sciences ,030218 nuclear medicine & medical imaging ,0104 chemical sciences ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Therapeutic index ,Oncology ,chemistry ,In vivo ,Radionuclide therapy ,Cancer research ,Medicine ,business ,medicine.drug - Abstract
Objective: The peptide receptor radionuclide therapy (PPRT) for somatostatin receptor positive (SSTR) neuroendocrine tumors (NETs) has emerged more than 15 years ago. The beta-emitter-PRRT has shown to induce objective response in 30-45% of metastatic NETs patients with hematologic/renal toxicity reduced by dose fractionation. The complete response to therapy is rare due to the heterogeneity of NETs; advanced stage of disease at the time of diagnosis; and patient resistance to nonradioactive octreotide and 90Y/177Lu PRRT developed during the therapy. The targeted alpha-emitter therapy of NETs can overcome these limitations. It can enhance the therapeutic response of patients and decrease side-effect and overcome patient resistance of beta-emitter PRRT without significant acute and mid-term toxicity. The RadioMedix and AREVA Med teams together have recently developed several novel 203Pb-peptide derivatives targeting SSTR(+) cancer cells, 203Pb-AR-RMX. The 203Pb is a gamma emitter (279 keV) with t1/2=51.9 h, suitable for single-photon emission computed tomography (SPECT) imaging. The 203Pb is an ideal surrogate for 212Pb α-particle therapy because both isotopes share identical chemical properties. The objective of these studies were: (1) to evaluate the SSTR targeting properties of novel conjugates 203Pb-AR-RMX; and (2) to determine their PK and biodistribution in vivo in SSTR overexpressing xenographs; and (3) to select lead candidate for further pre-IND clinical studies. Methods: AR-RMX was manufactured under GMP by Macrocylics Inc. The 203Pb-radiolabeling of AR-RMX was carried out under mild conditions. The SSTR targeting properties of 203Pb-AR-RMX were determined in the cellular uptake/competition studies in AR-42J cancer cells and in vivo in SSTR(+) AR42J xenograph mice. Results: 203Pb-AR-RMX-15 shows exceptionally high tumor-specific accumulation and retention in SSTR(+) AR42J xenograph mice. Tumor uptake of 203Pb-AR-RMX-15 was >14.4 %ID/g at 1h post injection and it remained at this level at least for 24h. The kidneys accumulation of 203Pb-AR-RMX-15 was >13 %ID/g at 1h and varies in different strains of mice but decreased progressively over the 24h. The kidney uptake of agent is similar to previously observed for octreotate labeled with other isotopes. The tumor uptake is significantly higher possible as a result of the change of charge of the 203Pb-AR-RMX-15. The acute hematotoxicity and chronic kidney toxicity is known to limit the doses of 90Y/177Lu PRRT. Preliminary studies of 203Pb-AR-RMX-15 showed that its kidney retention can be reduced by >32% by co-injection of our proprietary AminoMedixTM or Gelofusine-Lysine composition. 203Pb-AR-RMX-15 has shown >98% radio/chemical stability up 7 days post-formulation; no bone marrow uptake of agent was observed in bioD studies of 203Pb AR-RMX-15 done up to 24h post injection. These results allow us to hypothesize that the therapeutic dose of 203Pb/212Pb-AR-RMX-15 is expected to be significantly higher than the dose limiting activity. Conclusions: 203Pb-AR-RMX-15 showed promising results in vitro and in vivo studies and will be further investigated as a 203Pb/212Pb-labeled theranostics agent. Our “theranostics” approach using 203Pb/212Pb-PRRT has a potential to advance image-guided radionuclide therapy that can detect and deliver therapeutic radiation dose precisely to SSTR(+) NETs. Citation Format: Izabela Tworowska, Tania Stallons, Amal Saidi, Nilesh Wagh, Federico Rojas-Quijano, Paul Jurek, Garry Kiefer, Julien Torgue, Ebrahim Delpassand. Pb203-AR-RMX conjugates for image-guided TAT of neuroendocrine tumors (NETs) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr LB-259. doi:10.1158/1538-7445.AM2017-LB-259
- Published
- 2017
- Full Text
- View/download PDF
15. Detection of Canonical Hedgehog Signaling in Breast Cancer by 131-Iodine-Labeled Derivatives of the Sonic Hedgehog Protein
- Author
-
David J. Yang, Izabela Tworowska, Richard Larson, Wendy A. Woodward, Lynn M. Opdenaker, Ning Tsao, Firas Mourtada, Jennifer Sims-Mourtada, and Daniel L. Smith
- Subjects
Health, Toxicology and Mutagenesis ,Receptor expression ,lcsh:Medicine ,Iodine Radioisotopes ,chemistry.chemical_compound ,Tissue Distribution ,Receptor ,Oncogene Proteins ,Veratrum Alkaloids ,General Medicine ,Hedgehog signaling pathway ,Patched-1 Receptor ,Isotope Labeling ,embryonic structures ,Molecular Medicine ,Biological Assay ,Female ,Signal transduction ,Biotechnology ,Research Article ,Protein Binding ,Signal Transduction ,Patched ,Patched Receptors ,medicine.medical_specialty ,animal structures ,Article Subject ,Cyclopamine ,lcsh:Biotechnology ,Blotting, Western ,Breast Neoplasms ,Receptors, Cell Surface ,Biology ,Zinc Finger Protein GLI1 ,Internal medicine ,lcsh:TP248.13-248.65 ,Cell Line, Tumor ,Genetics ,medicine ,Sonic Hedgehog Protein ,Animals ,Humans ,Gamma Cameras ,Hedgehog Proteins ,Radiometry ,Radionuclide Imaging ,Molecular Biology ,Hedgehog ,Cell Proliferation ,lcsh:R ,Xenograft Model Antitumor Assays ,Rats, Inbred F344 ,Rats ,Endocrinology ,chemistry ,Cancer research ,Trans-Activators - Abstract
Activation of hedgehog (HH) pathway signaling is observed in many tumors. Due to a feedback loop, the HH receptor Patched (PTCH-1) is overexpressed in tumors with activated HH signaling. Therefore, we sought to radiolabel the PTCH-1 ligand sonic (SHH) for detection of cancer cells with canonical HH activity. Receptor binding of131I-SHH was increased in cell lines with high HH pathway activation. Our findings also show that PTCH-1 receptor expression is decreased upon treatment with HH signaling inhibitors, and receptor binding of131I-SHH is significantly decreased following treatment with cyclopamine.In vivoimaging and biodistribution studies revealed significant accumulation of131I-SHH within tumor tissue as compared to normal organs. Tumor-to-muscle ratios were approximately 8 : 1 at 5 hours, while tumor to blood and tumor to bone were 2 : 1 and 5 : 1, respectively. Significant uptake was also observed in liver and gastrointestinal tissue. These studies show that131I-SHH is capable ofin vivodetection of breast tumors with high HH signaling. We further demonstrate that the hedgehog receptor PTCH-1 is downregulated upon treatment with hedgehog inhibitors. Our data suggests that radiolabeled SHH derivatives may provide a method to determine response to SHH-targeted therapies.
- Published
- 2012
16. Base pairing within the psi32,psi39-modified anticodon arm of Escherichia coli tRNA(Phe)
- Author
-
Edward P. Nikonowicz and Izabela Tworowska
- Subjects
Magnetic Resonance Spectroscopy ,Base pair ,Stereochemistry ,Crystal structure ,medicine.disease_cause ,Biochemistry ,Catalysis ,RNA, Transfer, Phe ,Colloid and Surface Chemistry ,medicine ,Anticodon ,Escherichia coli ,Nucleotide ,Base Pairing ,Intramolecular Transferases ,chemistry.chemical_classification ,Base Sequence ,Hydrogen bond ,Chemistry ,Hydrogen Bonding ,General Chemistry ,Nuclear magnetic resonance spectroscopy ,Heteronuclear molecule ,Transfer RNA ,Nucleic Acid Conformation ,Ribosomes ,Pseudouridine - Abstract
The base-base hydrogen bond interactions of the psi32,psi39-modified anticodon arm of Escherichia coli tRNAPhe have been investigated using heteronuclear NMR spectroscopy. psi32 and psi39 were enzymatically introduced into a [13C,15N]-isotopically enriched RNA sequence corresponding to the tRNAPhe anticodon arm. Both the psi32-A38 and A31-psi39 nucleotide pairs form Watson-Crick base pairing schemes and the anticodon nucleotides adopt a triloop conformation. Similar effects were observed previously with D2-isopentenyl modification of the A37 N6 that also is native to the tRNAPhe anticodon arm. These results demonstrate that the individual modifications are not sufficient to produce the 32-38 bifurcated hydrogen bond or the U-turn motifs that are observed in crystal structures of tRNAs and tRNA-protein complexes. Thus the formation of these conserved structural features in solution likely require the synergistic interaction of multiple modifications.
- Published
- 2006
17. Long-term survival and toxicity profile of patients with disseminated neuroendocrine tumors following multiple cycles of high activity 111In pentetreotide therapy
- Author
-
R. Sharif, Lowell Anthony, Ebrahim S. Delpassand, Izabela Tworowska, and Jennifer Sims-Mourtada
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,business.industry ,Neuroendocrine tumors ,Multiple dosing ,medicine.disease ,Internal medicine ,Long term survival ,medicine ,111In-Pentetreotide ,High activity ,In patient ,business ,Toxicity profile - Abstract
e13545 Background: We have previously reported clinical response to multiple doses of high activity 111In pentetreotide in patients with disseminated neuroendocrine tumors. Here we report long term...
- Published
- 2010
- Full Text
- View/download PDF
18. Abstract 4335: Imaging of cancer stem cells with radiolabeled Hedgehog
- Author
-
Wendy A. Woodward, Firas Mourtada, Jennifer Sims-Mourtada, Jessica Li, and Izabela Tworowska
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Receptor expression ,Cancer ,Biology ,medicine.disease ,Embryonic stem cell ,medicine.anatomical_structure ,Oncology ,Cancer stem cell ,Cancer cell ,medicine ,Cancer research ,Bone marrow ,Progenitor cell ,Stem cell - Abstract
Recent studies have shown that the majority of early disseminated cancer cells detected in the bone marrow of breast cancer patients have a putative breast cancer “stem cell-like” phenotype. Moreover, activation of stem cell pathways in cancer cells has also been associated with resistance to radiation and chemotherapy. Early detection of these micrometastatic “stem cell-like” cancer cells could allow for earlier commencement of aggressive treatment to minimize relapse and disease spread and to improve therapeutic outcome. However, there is currently no stem cell targeted method to detect this population in vivo. New imaging agents are needed for real-time, non-invasive detection of stem cell pathway activation in micrometastatic disease in breast cancer. The stem cell pathway Hedgehog (HH) is known to regulate cell cycle entry and proliferation of embryonic neural and mammary progenitor cells as well as self-renewal of malignant mammary stem cells. Furthermore, the hedgehog receptor PTCH is overexpressed on cancer stem cells compared to normal mammary stem cells. Therefore, we sought to radiolabel the natural hedgehog ligand, sonic (SHH) with the positron emitter Gallium 68 (68Ga) for detection of cancer stem cells by positron emission tomography (PET). Recombinant SHH ligand was conjugated to the bifunctional chelator DOTA. DOTA-SHH was obtained with a total yield of 45%-60% as assessed by HPLC. DOTA-SHH was further characterized using MALDI-MS, ESI-MS with purity > 90%. The conjugate was radiolabeled with 68Ga, with radiochemical purity of >98% and specific activity of 4.3E+05 MBq/gram (theoretical specific activity = 9.18E+09 MBq/gram) as assessed by ITLC and radioHPLC. 68Ga-DOTA-SHH maintains high receptor binding in breast cancer cells and cellular uptake was correlated with PTCH receptor expression levels. Addition of 100-fold excess cold SHH reduced cellular uptake by approximately 90%. PTCH receptor expression is increased in MCF-7 stem cell-enriching culture conditions (mammospheres) and cellular uptake of 68Ga-DOTA-SHH was approximately 12-fold higher in mammosphere MCF-7 cultures compared to the total cell population. These findings demonstrate that chemical modification and radiolabeling of SHH ligand is feasible. Our studies suggest that 68Ga-DOTA-SHH has potential for molecular imaging of breast cancer by PET. Additionally, our studies suggest that 68Ga-DOTA-SHH can potentially identify aggressive tumors that may be rich in cancer stem cells. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4335.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.