1
|
West M, Flores JM, Mulhall JP. Contemporary prostate cancer radiation therapy trials may underestimate the risk of biochemical recurrence. J Sex Med 2025; 22:369-372. [PMID: 39786493 DOI: 10.1093/jsxmed/qdae191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/29/2024] [Accepted: 12/12/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Radiotherapy is often given with androgen deprivation therapy for prostate cancer which causes a reduction in testosterone levels, which when below castrate levels, can cause the prostate specific antigen (PSA) levels to be artificially low. AIM To determine if high-level radiotherapy clinical trials are underestimating biochemical recurrence (BCR) rates due to inadequate measurement of testosterone levels. METHODS The study plans for clinical trials performed by the Radiation Therapy Oncology Group (RTOG [now NRG]) on clinicaltrials.gov were reviewed for details on testosterone measurement in trials from 1994 to 2023, namely if the testosterone levels were indexed to PSA levels. OUTCOMES PSA being indexed to testosterone levels and other metrics of testosterone measurement, including time of day of measurement, assay used, and mean testosterone measurement. RESULTS Five of 21 (24%) trials stipulated that testosterone levels should be indexed to PSA levels. Eleven of 21 (52%) trials made no mention of testosterone. No trial reported testosterone assay or time of day of measurement. Thirteen of 21 (62%) trials did not require regular follow-up testosterone measurements. CLINICAL IMPLICATIONS The number of clinical trials indexing or regularly measuring testosterone was surprisingly low, which could cause an underestimation of BCR rates. STRENGTHS AND LIMITATIONS Strengths include being the first study, to our knowledge, to analyze the details of testosterone measurement in high-level radiotherapy trials. Limitations include only analyzing RTOG/NRG trials, analyzing unpublished data, and using clinicaltrials.gov rather than official trial protocols to determine details of testosterone measurement. CONCLUSION Indexing of testosterone levels to PSA levels in high-level radiotherapy trials using androgen deprivation therapy was uncommon, possibly rendering data on BCR unreliable, potentially underestimating BCR rates.
Collapse
Affiliation(s)
- Michael West
- Sexual & Reproductive Medicine Program, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Jose M Flores
- Sexual & Reproductive Medicine Program, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - John P Mulhall
- Sexual & Reproductive Medicine Program, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| |
Collapse
|
2
|
Laguna J, Wijngaard R, Hidalgo S, González-Escribano C, Ortiz V, Bedini JL, Filella X. Asociación entre la 25-hidroxivitamina D y el antígeno prostático específico: un estudio retrospectivo en hombres sin patologías prostáticas. ADVANCES IN LABORATORY MEDICINE 2023; 4:413-418. [PMID: 38106496 PMCID: PMC10724857 DOI: 10.1515/almed-2023-0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/01/2023] [Indexed: 12/19/2023]
Abstract
Objetivos Aunque estudios recientes asocian la vitamina D con el cáncer de próstata, otros estudios descartan una asociación entre esta vitamina y el cáncer de próstata o el antígeno prostático específico (PSA). Dado que no se pueden extraer conclusiones de los datos existentes, realizamos un estudio para analizar la relación entre el PSA y la 25-hidroxivitamina D [25(OH)D]. Métodos Un total de 415 sujetos sin patologías prostáticas fueron seleccionados, y se clasificaron por edad y concentraciones de 25(OH)D. El análisis estadístico se realizó con la prueba de Shapiro-Wilk, la prueba t de Student, ANOVA, y el coeficiente de correlación de Pearson. Además, se calculó el tamaño mínimo de muestra requerido para obtener resultados estadísticamente significativos en función de la concentración de 25(OH)D. Así mismo, se realizó la prueba t de Student para muestras pareadas para analizar a los individuos con dos determinaciones de PSA espaciadas en el tiempo en los que las concentraciones de 25(OH)D aumentaron o disminuyeron más de un 25 %. Resultados Observamos una leve correlación entre la edad y el PSA (r=0,379, p<0,001). Sin embargo, al comparar la concentración de PSA entre grupos en función de 25(OH)D, no se hallaron diferencias significativas (p=0,891): 1,25±1,32 μg/L (grupo con 25(OH)D<50 nmol/L) y 1,17±0,90 (grupo con 25(OH)D≥50 nmol/L). El coeficiente de correlación de Pearson fue casi 0. El tamaño mínimo de la muestra necesario para obtener resultados estadísticamente significativos fue de 815.346 hombres. No observamos diferencias en las concentraciones de PSA en los individuos que se sometieron a dos determinaciones. Conclusiones Nuestros resultados muestran que no existe asociación entre los niveles de 25(OH)D y de PSA en hombres sin patologías prostáticas.
Collapse
Affiliation(s)
- Javier Laguna
- Servicio de Bioquímica y Genética Molecular, CDB, Hospital Clínic de Barcelona, Barcelona, España
| | - Robin Wijngaard
- Servicio de Bioquímica y Genética Molecular, CDB, Hospital Clínic de Barcelona, Barcelona, España
| | - Susana Hidalgo
- Servicio de Bioquímica y Genética Molecular, CDB, Hospital Clínic de Barcelona, Barcelona, España
| | | | - Victoria Ortiz
- Laboratorio CORE, CDB, Hospital Clínic de Barcelona, BarcelonaEspaña
| | - José Luis Bedini
- Servicio de Bioquímica y Genética Molecular, CDB, Hospital Clínic de Barcelona, Barcelona, España
- Laboratorio CORE, CDB, Hospital Clínic de Barcelona, BarcelonaEspaña
| | - Xavier Filella
- Servicio de Bioquímica y Genética Molecular, CDB, Hospital Clínic de Barcelona, Barcelona, España
| |
Collapse
|
3
|
Laguna J, Wijngaard R, Hidalgo S, González-Escribano C, Ortiz V, Bedini JL, Filella X. Association between serum 25-hydroxyvitamin D and prostate-specific antigen: a retrospective study in men without prostate pathology. ADVANCES IN LABORATORY MEDICINE 2023; 4:408-412. [PMID: 38106489 PMCID: PMC10724855 DOI: 10.1515/almed-2023-0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/01/2023] [Indexed: 12/19/2023]
Abstract
Objectives Recently, vitamin D status has been associated with prostate cancer risk. However, some studies argue that there is no association of vitamin D with prostate cancer risk and serum prostate-specific antigen (PSA) concentrations. No clear conclusions can be drawn from the studies found in the literature. Our aim was to assess the relationship between PSA and 25-hydroxyvitamin D [25(OH)D]. Methods We selected 415 individuals without prostate pathologies and subgroups were generated according to age and 25(OH)D. Statistical analyses were performed using Shapiro-Wilk test, Student's t and ANOVA tests, and Pearson's correlation. Besides, the minimum sample size needed to obtain statistically significant results between groups according to 25(OH)D concentration was calculated and a Student's t-test for paired samples was performed to study individuals with two PSA measurements over time, where 25(OH)D concentration increased or decreased more than 25 %. Results We observed a slight correlation between age and PSA concentration (r=0.379, p<0.001). However, we found no significant differences when we compared PSA concentrations between groups according to 25(OH)D concentrations (p=0.891): 1.25 ± 1.32 μg/L (group with 25(OH)D<50 nmol/L) and 1.17 ± 0.90 (group with 25(OH)D≥50 nmol/L). Pearson's correlation coefficient was close to 0. The minimum samples size to obtain statistically significant results was 815,346 men, and we observed no differences in PSA concentrations in individuals with two measurements. Conclusions Our findings show no association in men without prostate pathologies, based on 25(OH)D levels.
Collapse
Affiliation(s)
- Javier Laguna
- Department of Biochemistry and Molecular Genetics, CDB, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Robin Wijngaard
- Department of Biochemistry and Molecular Genetics, CDB, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Susana Hidalgo
- Department of Biochemistry and Molecular Genetics, CDB, Hospital Clínic of Barcelona, Barcelona, Spain
| | | | - Victoria Ortiz
- CORE Laboratory, CDB, Hospital Clínic of Barcelona, Barcelona, Spain
| | - José Luis Bedini
- Department of Biochemistry and Molecular Genetics, CDB, Hospital Clínic of Barcelona, Barcelona, Spain
- CORE Laboratory, CDB, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Xavier Filella
- Department of Biochemistry and Molecular Genetics, CDB, Hospital Clínic of Barcelona, Barcelona, Spain
| |
Collapse
|
4
|
Hoareau R, Bach-Gansmo T, Cumming P, Olberg DE. A new automated and putatively versatile synthesis of the PSMA-ligand derivative [ 18F]DCFPyL using the FASTlab TM synthesizer. EJNMMI Radiopharm Chem 2022; 7:10. [PMID: 35507241 PMCID: PMC9068851 DOI: 10.1186/s41181-022-00157-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/03/2022] [Indexed: 11/24/2022] Open
Abstract
Background Noninvasive molecular imaging using peptides and biomolecules labelled with positron emitters has become important for detection of cancer and other diseases with PET (positron emission tomography). The positron emitting radionuclide fluorine-18 is widely available in high yield from cyclotrons and has favorable decay (t1/2 109.7 min) and imaging properties. 18F-Labelling of biomolecules and peptides for use as radiotracers is customarily achieved in a two-step approach, which can be challenging to automate. 6-[18F]Fluoronicotinic acid 2,3,5,6-tetrafluorophenyl ester ([18F]F-Py-TFP) is a versatile 18F-prosthetic group for this purpose, which can be rapidly be produced in an one-step approach on solid support. This work details an automated procedure on the cassette-based GE FASTlab™ platform for the labeling of a peptidomimetic, exemplified by the case of using the Glu-CO-Lys motif to produce [18F]DCFPyL, a ligand targeting the prostate specific membrane antigen (PSMA). Results From fluorine-18 delivery a fully automated two-step radiosynthesis of [18F]DCFPyL was completed in 56 min with an overall end of synthesis yield as high as 37% using solid phase extraction (SPE) purification on the GE FASTlab™ platform. Conclusions Putatively, this radiolabeling methodology is inherently amenable to automation with a diverse set of synthesis modules, and it should generalize for production of a broad spectrum of biomolecule-based radiotracers for use in PET imaging.
Collapse
Affiliation(s)
| | | | - Paul Cumming
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, Australia.,Institute of Nuclear Medicine, Inelspital, University of Bern, Bern, Switzerland
| | - Dag Erlend Olberg
- Norsk Medisinsk Syklotronsenter AS, Postboks 4950, 0424, Nydalen, Oslo, Norway. .,School of Pharmacy, University of Oslo, Oslo, Norway.
| |
Collapse
|
5
|
Flores JM, Bernie HL, Miranda E, Nascimento B, Schofield E, Benfante N, Carlsson S, Mulhall JP. The Relationship Between PSA and Total Testosterone Levels in Men With Prostate Cancer. J Sex Med 2022; 19:471-478. [PMID: 35135736 PMCID: PMC9359436 DOI: 10.1016/j.jsxm.2022.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/10/2021] [Accepted: 01/05/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND Prostate-specific antigen (PSA) secretion is a testosterone (T) dependent process. Published data suggest that a low T level is an independent predictor of higher-grade prostate cancer (PC). AIM To evaluate the relationship between T and PSA in patients with PC. METHODS All men diagnosed with PC with a recorded pre-treatment total T level measurement were included in this analysis. We analyzed demographic, clinical, and pathological data. Patients were stratified according to pretreatment PSA levels: <2 ng/mL, 2-4 ng/mL, >4 ng/mL. Low T was defined as total T < 10.4 nmol/L (300 ng/dL), very low T < 6.9 nmol/L (200 ng/dL). OUTCOMES T levels by PSA groups according to the PC pathology. RESULTS In this retrospective study, mean patient age was 61 years among 646 men. The distribution by PSA group was: 8% (<2), 17% (2-4), and 76% (>4). The mean T level across the entire cohort was 13 nmol/L (374 ng/dL). Overall, 30% had a T level < 10.4 nmol/L (300 ng/dL). The mean total T level by PSA group was: <2 ng/mL, 7 nmol/L (206 ng/dL); 2-4 ng/mL, 13 nmol/L (362 ng/dL); >4 ng/mL, 14 nmol/L (393 ng/dL), P < .001. PSA <4 ng/mL was a significant predictor of low T in men with PC GS ≥8. PSA <2 ng/mL was a significant predictor of very low T independent of the PC pathology. CLINICAL IMPLICATIONS These findings suggest that clinicians should consider measuring T levels when a patient diagnosed with PC GS ≥8 and PSA level <4 ng/mL, and for each patient with PSA level <2 ng/mL independent of the PC pathology. STRENGTHS & LIMITATIONS Our study has several strengths including (i) inclusion of a large population of men, (ii) use of a database which is audited and reviewed for accuracy annually, and (iii) use of an accurate T assay (LCMS). Nonetheless, there are limitations: (i) the subjects of the study are from a single institution, and (ii) we did not measure free T levels. CONCLUSION In men with PC with GS ≥8, PSA level <4 ng/mL predicts low T. PSA <2 ng/mL predicts very low T independent of the PC pathology. Flores JM, Bernie HL, Miranda E, et al. The Relationship Between PSA and Total Testosterone Levels in Men With Prostate Cancer. J Sex Med 2022;19:471-478.
Collapse
Affiliation(s)
- Jose M Flores
- Sexual & Reproductive Medicine Program, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Helen L Bernie
- Sexual & Reproductive Medicine Program, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Eduardo Miranda
- Sexual & Reproductive Medicine Program, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Bruno Nascimento
- Sexual & Reproductive Medicine Program, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Elizabeth Schofield
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicole Benfante
- Sexual & Reproductive Medicine Program, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Sigrid Carlsson
- Urology Service, Department of Surgery, and Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - John P Mulhall
- Sexual & Reproductive Medicine Program, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY USA.
| |
Collapse
|
6
|
Wang Z, Wen P, Hu B, Cao S, Shi X, Guo W, Zhang S. Dopamine and dopamine receptor D1 as a novel favourable biomarker for hepatocellular carcinoma. Cancer Cell Int 2021; 21:586. [PMID: 34717619 PMCID: PMC8557590 DOI: 10.1186/s12935-021-02298-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/24/2021] [Indexed: 12/15/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) remains one of the most common malignant tumours worldwide. Therefore, the identification and development of sensitivity- genes as novel diagnostic markers and effective therapeutic targets is urgently needed. Dopamine and dopamine receptor D1 (DRD1) are reported to be involved in the progression of various cancers. However, the crucial role of DRD1 in HCC malignant activities remains unclear. Methods We enrolled 371 patients with liver hepatocellular carcinoma (LIHC) from The Cancer Genome Atlas (TCGA) to detect the expression and functions of DRD1. The Tumour Immune Estimation Resource (TIMER), UALCAN database, Kaplan–Meier plotter, cBioPortal database, and LinkedOmics database were utilized for the systematic investigation of DRD1 expression and related clinical features, coexpressed genes, functional pathways, mutations, and immune infiltrates in HCC. Results In this study, we determined that DRD1 expression was decreased in HCC tumour tissues versus normal tissues and that low DRD1 expression indicated a poor prognosis. The significance of DRD1 expression varied among different tumour samples. The somatic mutation frequency of DRD1 in the LIHC cohort was 0.3%. The biological functions of DRD1 were detected and validated, and DRD1 was shown to be involved in various functional activities, including metabolism, oxidation, mitochondrial matrix-related processes and other related signaling pathways. In addition, out study indicated that DRD1 had significant correlations with the infiltration of macrophages, B cells and CD+ T cells in HCC. Conclusions These findings demonstrated the rationality of the potential application of DRD1 function as a novel biomarker for HCC diagnosis and a therapeutic target for HCC treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02298-9.
Collapse
Affiliation(s)
- Zhihui Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Road, Zhengzhou, 450052, Henan, China. .,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China. .,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China. .,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China.
| | - Peihao Wen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Road, Zhengzhou, 450052, Henan, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China
| | - Bowen Hu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Road, Zhengzhou, 450052, Henan, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China
| | - Shengli Cao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Road, Zhengzhou, 450052, Henan, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China
| | - Xiaoyi Shi
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Road, Zhengzhou, 450052, Henan, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Road, Zhengzhou, 450052, Henan, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Road, Zhengzhou, 450052, Henan, China. .,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China. .,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China. .,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China.
| |
Collapse
|
7
|
Paller CJ, Piana D, Eshleman JR, Riel S, Denmeade SR, Velho PI, Rowe SP, Pomper MG, Antonarakis ES, Luo J, Eisenberger MA. A pilot study of prostate-specific membrane antigen (PSMA) dynamics in men undergoing treatment for advanced prostate cancer. Prostate 2019; 79:1597-1603. [PMID: 31361358 PMCID: PMC6818502 DOI: 10.1002/pros.23883] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 06/24/2019] [Indexed: 11/06/2022]
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA) is a rational target for noninvasive detection of recurrent prostate cancer (PCa) and for therapy of metastatic castration-resistant prostate cancer (mCRPC) with PSMA-targeted agents. Here we conducted serial measurements of PSMA expression on circulating tumor cells (CTCs) to evaluate patterns of longitudinal PSMA dynamics over the course of multiple sequential therapies. METHODS A retrospective investigation of men with mCRPC undergoing evaluation at medical oncology clinics at our institution assessed the dynamics of PSMA expression in the context of different systemic treatments administered sequentially. Eligibility included patients who began systemic therapies with androgen receptor (AR)-directed agents or taxane agents for whom peripheral blood samples were tested for CTC mRNA of AR splice variant-7 (AR-V7), prostate-specific antigen (PSA), and PSMA (with >2 CTC + results) in a CLIA-accredited laboratory. RESULTS From August 2015 to November 2017, we identified 96 eligible men. Fifteen had greater than or equal to 2 sequential therapies and evaluable CTC samples, greater than or equal to 1 expressing PSMA (PSMA+). Among the 15 patients included in this analysis, a total of 54 PSMA status evaluations were performed in the context of 48 therapies during a median follow-up of 18 months. At baseline, PSMA signal was detected ("positive") in 11 of 15 (73.3%) patients, while for 4 of 15 (26.7%) patients PSMA signal was undetectable ("negative"). In all but two patients, the baseline collection corresponded with a change in treatment. On the second assessment, PSMA increases were detected in all 4/4 (100%) PSMA-negative patients and 8 of 11 (72.7%) PSMA-positive patients. PSMA significantly decreased in a patient treated with 177 Lu-PSMA-617. Serum PSA declines were seen in 7 of 8 (88%) of the treatment periods where PSMA decreased. CONCLUSIONS PSMA expression in CTCs is a dynamic marker. PSMA transcript declines appear to be associated with concurrent decreases in serum PSA. Sequential CTC sampling could provide a noninvasive response assessment to systemic treatment for mCRPC.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Antigens, Surface/blood
- Antigens, Surface/genetics
- Bridged-Ring Compounds/therapeutic use
- Dipeptides/therapeutic use
- Glutamate Carboxypeptidase II/blood
- Glutamate Carboxypeptidase II/genetics
- Heterocyclic Compounds, 1-Ring/therapeutic use
- Humans
- Lutetium
- Male
- Middle Aged
- Neoplasm Recurrence, Local/blood
- Neoplasm Recurrence, Local/therapy
- Neoplastic Cells, Circulating/chemistry
- Pilot Projects
- Prostate-Specific Antigen/blood
- Prostate-Specific Antigen/genetics
- Prostatic Neoplasms/blood
- Prostatic Neoplasms/therapy
- Prostatic Neoplasms, Castration-Resistant/blood
- Prostatic Neoplasms, Castration-Resistant/therapy
- RNA, Messenger/blood
- Receptors, Androgen/drug effects
- Retrospective Studies
- Taxoids/therapeutic use
- Treatment Outcome
Collapse
Affiliation(s)
- Channing J. Paller
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Danilo Piana
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - James R. Eshleman
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutes, Baltimore, Maryland
| | - Stacy Riel
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutes, Baltimore, Maryland
| | - Samuel R. Denmeade
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Pedro Isaacsson Velho
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Steven P. Rowe
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Martin G. Pomper
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Emmanuel S. Antonarakis
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jun Luo
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mario A. Eisenberger
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
8
|
Collier TL, Maresca KP, Normandin MD, Richardson P, McCarthy TJ, Liang SH, Waterhouse RN, Vasdev N. Brain Penetration of the ROS1/ALK Inhibitor Lorlatinib Confirmed by PET. Mol Imaging 2018; 16:1536012117736669. [PMID: 29067878 PMCID: PMC5661750 DOI: 10.1177/1536012117736669] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The Massachusetts General Hospital Radiochemistry Program, in collaboration with Pfizer, has developed unique 11C and 18F-labeling strategies to synthesize isotopologs of lorlatinib (PF-06463922) which is undergoing phase III clinical trial investigations for treatment of non-small-cell lung cancers with specific molecular alterations. A major goal in cancer therapeutics is to measure the concentrations of this drug in the brain metastases of patients with lung cancer, and penetration of the blood–brain barrier is important for optimal therapeutic outcomes. Our recent publication in Nature Communications employed radiolabeled lorlatinib and positron emission tomography (PET) studies in preclinical models including nonhuman primates (NHPs) that demonstrated high brain permeability of this compound. Our future work with radiolabeled lorlatinib will include advanced PET evaluations in rodent tumor models and normal NHPs with the goal of clinical translation.
Collapse
Affiliation(s)
- T Lee Collier
- 1 Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital (MGH) & Department of Radiology, Harvard Medical School, Boston, MA, USA.,2 Advion, Inc, Ithaca, NY, USA
| | - Kevin P Maresca
- 3 Clinical and Translational Imaging, Worldwide Research and Development, Pfizer Inc, Cambridge, MA, USA
| | - Marc D Normandin
- 1 Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital (MGH) & Department of Radiology, Harvard Medical School, Boston, MA, USA
| | | | - Timothy J McCarthy
- 3 Clinical and Translational Imaging, Worldwide Research and Development, Pfizer Inc, Cambridge, MA, USA
| | - Steven H Liang
- 1 Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital (MGH) & Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Rikki N Waterhouse
- 3 Clinical and Translational Imaging, Worldwide Research and Development, Pfizer Inc, Cambridge, MA, USA.,5 Waterhouse Imaging and Biomarker Consultants, Chester, NH, USA
| | - Neil Vasdev
- 1 Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital (MGH) & Department of Radiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Molecular Imaging-Guided Radiotherapy for the Treatment of Head-and-Neck Squamous Cell Carcinoma: Does it Fulfill the Promises? Semin Radiat Oncol 2018; 28:35-45. [PMID: 29173754 DOI: 10.1016/j.semradonc.2017.08.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
With the routine use of intensity modulated radiation therapy for the treatment of head-and-neck squamous cell carcinoma allowing highly conformed dose distribution, there is an increasing need for refining both the selection and the delineation of gross tumor volumes (GTV). In this framework, molecular imaging with positron emission tomography and magnetic resonance imaging offers the opportunity to improve diagnostic accuracy and to integrate tumor biology mainly related to the assessment of tumor cell density, tumor hypoxia, and tumor proliferation into the treatment planning equation. Such integration, however, requires a deep comprehension of the technical and methodological issues related to image acquisition, reconstruction, and segmentation. Until now, molecular imaging has had a limited value for the selection of nodal GTV, but there are increasing evidences that both FDG positron emission tomography and diffusion-weighted magnetic resonance imaging has a potential value for the delineation of the primary tumor GTV, effecting on dose distribution. With the apprehension of the heterogeneity in tumor biology through molecular imaging, growing evidences have been collected over the years to support the concept of dose escalation/dose redistribution using a planned heterogeneous dose prescription, the so-called "dose painting" approach. Validation trials are ongoing, and in the coming years, one may expect to position the dose painting approach in the armamentarium for the treatment of patients with head-and-neck squamous cell carcinoma.
Collapse
|
10
|
Abstract
The process of discovering and developing a new pharmaceutical is a long, difficult, and risky process that requires numerous resources. Molecular imaging techniques such as PET have recently become a useful tool for making decisions along a drug candidate's development timeline. PET is a translational, noninvasive imaging technique that provides quantitative information about a potential drug candidate and its target at the molecular level. Using this technique provides decisional information to ensure that the right drug candidate is being chosen, for the right target, at the right dose within the right patient population. This review will focus on small molecule PET tracers and how they are used within the drug discovery process. PET provides key information about a drug candidate's pharmacokinetic and pharmacodynamic properties in both preclinical and clinical studies. PET is being used in all phases of the drug discovery and development process, and the goal of these studies are to accelerate the process in which drugs are developed.
Collapse
Affiliation(s)
- David J Donnelly
- Bristol-Myers Squibb Pharmaceutical Research and Development, Princeton, NJ.
| |
Collapse
|
11
|
Synthesis and preliminary PET imaging of 11C and 18F isotopologues of the ROS1/ALK inhibitor lorlatinib. Nat Commun 2017; 8:15761. [PMID: 28594000 PMCID: PMC5472746 DOI: 10.1038/ncomms15761] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/27/2017] [Indexed: 01/27/2023] Open
Abstract
Lorlatinib (PF-06463922) is a next-generation small-molecule inhibitor of the orphan receptor tyrosine kinase c-ros oncogene 1 (ROS1), which has a kinase domain that is physiologically related to anaplastic lymphoma kinase (ALK), and is undergoing Phase I/II clinical trial investigations for non-small cell lung cancers. An early goal is to measure the concentrations of this drug in brain tumour lesions of lung cancer patients, as penetration of the blood–brain barrier is important for optimal therapeutic outcomes. Here we prepare both 11C- and 18F-isotopologues of lorlatinib to determine the biodistribution and whole-body dosimetry assessments by positron emission tomography (PET). Non-traditional radiolabelling strategies are employed to enable an automated multistep 11C-labelling process and an iodonium ylide-based radiofluorination. Carbon-11-labelled lorlatinib is routinely prepared with good radiochemical yields and shows reasonable tumour uptake in rodents. PET imaging in non-human primates confirms that this radiotracer has high brain permeability. Lorlatinib—a ROS1/ALK inhibitor—is currently undergoing clinical trials for the treatment of non-small cell lung cancers. Here the authors develop synthetic routes to 11C- and 18F-labelled lorlatinib, with subsequent PET imaging showing good blood brain barrier permeability in non-human primates.
Collapse
|
12
|
Fowler AM, Clark AS, Katzenellenbogen JA, Linden HM, Dehdashti F. Imaging Diagnostic and Therapeutic Targets: Steroid Receptors in Breast Cancer. J Nucl Med 2016; 57 Suppl 1:75S-80S. [PMID: 26834106 DOI: 10.2967/jnumed.115.157933] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Estrogen receptor alpha (ERα) and progesterone receptor (PR) are important steroid hormone receptor biomarkers used to determine prognosis and to predict benefit from endocrine therapies for breast cancer patients. Receptor expression is routinely measured in biopsy specimens using immunohistochemistry, although such testing can be challenging, particularly in the setting of metastatic disease. ERα and PR can be quantitatively assayed noninvasively with PET. This approach provides the opportunity to assess receptor expression and function in real time, within the entire tumor, and across distant sites of metastatic disease. This article reviews the current evidence of ERα and PR PET imaging as predictive and early-response biomarkers for endocrine therapy.
Collapse
Affiliation(s)
- Amy M Fowler
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Amy S Clark
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Hannah M Linden
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, Washington; and
| | - Farrokh Dehdashti
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri
| |
Collapse
|
13
|
Boros E, Holland JP, Kenton N, Rotile N, Caravan P. Macrocycle-Based Hydroxamate Ligands for Complexation and Immunoconjugation of 89Zirconium for Positron Emission Tomography (PET) Imaging. Chempluschem 2016; 81:274-281. [PMID: 27630807 PMCID: PMC5019580 DOI: 10.1002/cplu.201600003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Indexed: 12/19/2022]
Abstract
Four novel chelators (L1-L4) and their 89zirconium complexes were prepared and compared with the 89zirconium desferrioxamine B (DFO) complex. The new chelates are based on 1,4,7,10-tetraazacyclododecane (cyclen) and 1,4,8,11-tetraazacyclotetradecane (cyclam) scaffolds and present either three or four hydroxamate arms for coordination with Zr4+ ions with coordination numbers between six and eight. The 89Zr-L4 complex showed similar stability to that of 89Zr-DFO when incubated in either rat blood plasma or ethylenediaminetetraacetic acid challenge experiments. Positron imaging and biodistribution studies in mice showed that 89Zr-L4 had similar pharmacokinetic behavior to that of 89Zr-DFO, with rapid renal elimination and low residual activity in background tissues. A bifunctional version of L4 (L5) was synthesized and conjugated to trastuzumab; an anti-HER2/neu antibody. Immunopositron emission tomography imaging and biodistribution with 89Zr-L5-trastuzumab revealed high tumor to background ratios (tumor/blood ratio: 14.2 ± 2.25) and a high tumor specificity that was comparable to the performance of 89Zr-DFO-trastuzumab.
Collapse
Affiliation(s)
- Eszter Boros
- The Athinoula A. Martinos Center for Biomedical Imaging Department of Radiology, Massachusetts General Hospital Harvard Medical School, 149 Thirteenth Street, Suite 2301 Charlestown, MA 02129 (USA)
| | - Jason P. Holland
- Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital, Department of Radiology Harvard Medical School, 55 Fruit Street, White 427 Boston, MA 02114 (USA)
| | - Nathaniel Kenton
- The Athinoula A. Martinos Center for Biomedical Imaging Department of Radiology, Massachusetts General Hospital Harvard Medical School, 149 Thirteenth Street, Suite 2301 Charlestown, MA 02129 (USA)
| | - Nicholas Rotile
- The Athinoula A. Martinos Center for Biomedical Imaging Department of Radiology, Massachusetts General Hospital Harvard Medical School, 149 Thirteenth Street, Suite 2301 Charlestown, MA 02129 (USA)
| | - Peter Caravan
- The Athinoula A. Martinos Center for Biomedical Imaging Department of Radiology, Massachusetts General Hospital Harvard Medical School, 149 Thirteenth Street, Suite 2301 Charlestown, MA 02129 (USA)
| |
Collapse
|
14
|
Gao L, Liu H, Sun X, Gao D, Zhang C, Jia B, Zhu Z, Wang F, Liu Z. Molecular Imaging of Post-Src Inhibition Tumor Signatures for Guiding Dasatinib Combination Therapy. J Nucl Med 2016; 57:321-326. [PMID: 26383149 DOI: 10.2967/jnumed.115.158881] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 08/26/2015] [Indexed: 01/27/2023] Open
Abstract
UNLABELLED Noninvasive, real-time, quantitative measurement of key biomarkers associated with cancer therapeutic interventions could provide a better understanding of cancer biology. We investigated in this study whether incorporating multiple molecular imaging approaches could be used to guide dasatinib anti-Src therapy and aid in the rational design of a combination therapy regimen. METHODS Bioluminescence imaging, (18)F-FDG PET, integrin αvβ3-targeted SPECT/CT, and vascular endothelial growth factor-targeted near-infrared fluorescence imaging were performed before and after dasatinib treatment in a tumor mouse model. RESULTS There was no significant difference in the bioluminescence imaging signal or (18)F-FDG tumor uptake in dasatinib-treated tumors compared with the control tumors. However, the uptake of (99m)T-3PRGD2 (integrin αvβ3-specific) and DyLight755-ranibizumab (vascular endothelial growth factor-specific) in the dasatinib-treated tumors was significantly lower than that in the control tumors. In vitro studies confirmed the antiangiogenic effects of dasatinib but indicated a lack of cytotoxicity. Dasatinib plus cytotoxic docetaxel elicited marked synergistic tumor growth inhibition in vivo. CONCLUSION Visualization of post-Src inhibition tumor signatures through multiple imaging approaches facilitates sensitive and quantitative measurement of cancer biomarkers in vivo, thus aiding in the rational design of dasatinib combination therapy.
Collapse
Affiliation(s)
- Liquan Gao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hao Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xianlei Sun
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Duo Gao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Chenran Zhang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Zhaohui Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China; and
| | - Fan Wang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China Interdisciplinary Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhaofei Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| |
Collapse
|
15
|
Image-derived biomarkers and multimodal imaging strategies for lung cancer management. Eur J Nucl Med Mol Imaging 2015; 42:634-43. [DOI: 10.1007/s00259-014-2974-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 12/08/2014] [Indexed: 12/14/2022]
|
16
|
Neal JW, Gainor JF, Shaw AT. Developing biomarker-specific end points in lung cancer clinical trials. Nat Rev Clin Oncol 2014; 12:135-46. [PMID: 25533947 DOI: 10.1038/nrclinonc.2014.222] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In cancer-drug development, a number of different end points have been used to establish efficacy and support regulatory approval, such as overall survival, progression-free survival (PFS), and radiographic response rate. However, these traditional end points have important limitations. For example, in lung cancer clinical trials, evaluating overall survival end points is a protracted process and these end points are most reliable when crossover to the investigational therapy is not permitted. Furthermore, although radiographic surrogate end points, such as PFS and response rate, generally correlate with clinical benefit in the setting of cytotoxic chemotherapy and molecular targeted therapies, novel immunotherapies might have atypical response kinetics, which confounds radiographic interpretation. In this Review, we discuss the need to develop alternative or surrogate end points for lung cancer clinical trials, and focus on several new biomarkers that could serve as surrogate end points, including functional imaging biomarkers, circulating factors (tumour proteins, DNA, and cells), and pharmacodynamic tumour markers. By enabling the size, duration, and complexity of cancer trials to be reduced, biomarker end points hold the promise to accelerate drug development and improve patient outcomes.
Collapse
Affiliation(s)
- Joel W Neal
- Department of Medicine, Division of Oncology, Stanford Cancer Institute and Stanford University School of Medicine, Stanford University, 875 Blake Wilbur Drive, Stanford, CA 94305, USA
| | - Justin F Gainor
- Division of Hematology-Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, 32 Fruit Street, Boston, MA 02114, USA
| | - Alice T Shaw
- Division of Hematology-Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, 32 Fruit Street, Boston, MA 02114, USA
| |
Collapse
|
17
|
Ma T, Sun X, Cui L, Gao L, Wu Y, Liu H, Zhu Z, Wang F, Liu Z. Molecular imaging reveals trastuzumab-induced epidermal growth factor receptor downregulation in vivo. J Nucl Med 2014; 55:1002-1007. [PMID: 24732154 DOI: 10.2967/jnumed.114.137000] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 03/11/2014] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Previous in vitro studies demonstrated that treating tumors expressing both epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 with trastuzumab resulted in increased EGFR homodimerization and subsequent rapid downregulation of EGFR. We investigated whether molecular imaging using near-infrared fluorescence (NIRF) imaging and PET probes could sensitively detect trastuzumab-induced EGFR downregulation in vivo. METHODS The F(ab')2 antibody fragment PaniF(ab')2 was generated by digesting the anti-EGFR monoclonal antibody panitumumab. PaniF(ab')2 was labeled with either a NIRF dye or (68)Ga, and optical imaging and small-animal PET imaging of Dye-PaniF(ab')2 and (68)Ga-PaniF(ab')2, respectively, were performed in HT-29 tumor-bearing nude mice treated with trastuzumab or untreated control. RESULTS Longitudinal NIRF imaging studies revealed significantly reduced tumor uptake of Dye-PaniF(ab')2 on days 5 and 7 in trastuzumab-treated HT-29 tumors, compared with control. Western blotting confirmed the downregulation of EGFR after treatment with trastuzumab. Small-animal PET on day 5 after trastuzumab treatment also demonstrated decreased (68)Ga-PaniF(ab')2 uptake in trastuzumab-treated HT-29 tumors. The tumor uptake value of (68)Ga-PaniF(ab')2 obtained from PET imaging had an excellent linear correlation with the uptake value measured using biodistribution. CONCLUSION The downregulation of EGFR induced by trastuzumab treatment could be detected noninvasively using optical and PET imaging. This molecular imaging strategy could provide a dynamic readout of changes in the tumor signaling and may facilitate the noninvasive monitoring of the early tumor response to drug treatment.
Collapse
Affiliation(s)
- Teng Ma
- Medical Isotopes Research Center, Peking University, Beijing, China Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and
| | - Xianlei Sun
- Medical Isotopes Research Center, Peking University, Beijing, China Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and
| | - Liyang Cui
- Medical Isotopes Research Center, Peking University, Beijing, China Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and
| | - Liquan Gao
- Medical Isotopes Research Center, Peking University, Beijing, China Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and
| | - Yue Wu
- Medical Isotopes Research Center, Peking University, Beijing, China Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and
| | - Hao Liu
- Medical Isotopes Research Center, Peking University, Beijing, China Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and
| | - Zhaohui Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Fan Wang
- Medical Isotopes Research Center, Peking University, Beijing, China Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and
| | - Zhaofei Liu
- Medical Isotopes Research Center, Peking University, Beijing, China Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and
| |
Collapse
|
18
|
Leimgruber A, Möller A, Everitt SJ, Chabrot M, Ball DL, Solomon B, MacManus M, Hicks RJ. Effect of Platinum-Based Chemoradiotherapy on Cellular Proliferation in Bone Marrow and Spleen, Estimated by (18)F-FLT PET/CT in Patients with Locally Advanced Non-Small Cell Lung Cancer. J Nucl Med 2014; 55:1075-80. [PMID: 24868108 DOI: 10.2967/jnumed.113.136127] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 03/07/2014] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Historically, it has been difficult to monitor the acute impact of anticancer therapies on hematopoietic organs on a whole-body scale. Deeper understanding of the effect of treatments on bone marrow would be of great potential value in the rational design of intensive treatment regimens. 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT) is a functional radiotracer used to study cellular proliferation. It is trapped in cells in proportion to thymidine-kinase 1 enzyme expression, which is upregulated during DNA synthesis. This study investigates the potential of (18)F-FLT to monitor acute effects of chemotherapy on cellular proliferation and its recovery in bone marrow, spleen, and liver during treatment with 2 different chemotherapy regimens. METHODS Sixty patients with non-small cell lung cancer underwent concurrent radical chemoradiotherapy to 60 Gy in 6 wk with either cisplatin/etoposide (C/E, n = 28) weeks 1 and 5 or weekly carboplatin/paclitaxel (C/P, n = 32) regimens. (18)F-FLT and (18)F-FDG PET with CT were performed at baseline, week 2 (day 9 for (18)F-FLT and day 10 for (18)F-FDG PET), and week 4 (day 23 for (18)F-FLT and day 24 for (18)F-FDG PET). Visual and semiquantitative standardized uptake value (SUV) measurements were performed in bone marrow outside the radiotherapy field, liver, spleen, and small bowel. These were correlated to blood counts and smears in a subset of patients. RESULTS The C/E group exhibited a drop in bone marrow (18)F-FLT uptake at week 2 (median SUVmax [maximum SUV] decrease to 31%, 8.7-6.0, P < 0.001), with recovery at week 4, reflecting the absence of chemotherapy between these times. By contrast, the weekly C/P group showed gradually declining bone marrow uptake (P > 0.05). Spleen uptake in both cohorts decreased at week 2, with intense rebound activity at week 4 (SUVmax week 4 at 58% above baseline: 2.4-3.8, for C/E, respectively, 30% for C/P: 2.7-3.5, P < 0.001). Liver uptake changed little. (18)F-FLT changes preceded neutrophil count reductions. (18)F-FDG uptake in marrow liver and spleen changed much less than (18)F-FLT. CONCLUSION (18)F-FLT imaging may be used to quantify impairment and recovery of bone marrow by specific chemotherapy regimens and may also enable imaging of organ-specific processes such as spleen activation. (18)F-FLT is superior to (18)F-FDG for this purpose. This technology may support novel treatment planning and monitoring approaches in oncology patients.
Collapse
Affiliation(s)
- Antoine Leimgruber
- Centre for Molecular Imaging, Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia Department of Medical Imaging, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Andreas Möller
- QIMR Berghofer Medical Research Institute, Herston Queensland, Australia
| | - Sarah J Everitt
- Department of Radiation Oncology, Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia Department of Medical Imaging and Radiation Sciences, Monash University, Clayton, Victoria, Australia; and
| | - Marine Chabrot
- Centre for Molecular Imaging, Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - David L Ball
- Department of Radiation Oncology, Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Ben Solomon
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia Medical Oncology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Michael MacManus
- Department of Radiation Oncology, Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Rodney J Hicks
- Centre for Molecular Imaging, Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
19
|
Adiseshaiah PP, Patel NL, Ileva LV, Kalen JD, Haines DC, McNeil SE. Longitudinal imaging of cancer cell metastases in two preclinical models: a correlation of noninvasive imaging to histopathology. INTERNATIONAL JOURNAL OF MOLECULAR IMAGING 2014; 2014:102702. [PMID: 24724022 PMCID: PMC3958723 DOI: 10.1155/2014/102702] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 01/18/2014] [Indexed: 01/24/2023]
Abstract
Metastatic spread is the leading cause of death from cancer. Early detection of cancer at primary and metastatic sites by noninvasive imaging modalities would be beneficial for both therapeutic intervention and disease management. Noninvasive imaging modalities such as bioluminescence (optical), positron emission tomography (PET)/X-ray computed tomography (CT), and magnetic resonance imaging (MRI) can provide complementary information and accurately measure tumor growth as confirmed by histopathology. Methods. We validated two metastatic tumor models, MDA-MD-231-Luc and B16-F10-Luc intravenously injected, and 4T1-Luc cells orthotopically implanted into the mammary fat pad. Longitudinal whole body bioluminescence imaging (BLI) evaluated metastasis, and tumor burden of the melanoma cell line (B16-F10-Luc) was correlated with (PET)/CT and MRI. In addition, ex vivo imaging evaluated metastasis in relevant organs and histopathological analysis was used to confirm imaging. Results. BLI revealed successful colonization of cancer cells in both metastatic tumor models over a 4-week period. Furthermore, lung metastasis of B16-F10-Luc cells imaged by PET/CT at week four showed a strong correlation (R (2) = 0.9) with histopathology. The presence and degree of metastasis as determined by imaging correlated (R (2) = 0.7) well with histopathology findings. Conclusions. We validated two metastatic tumor models by longitudinal noninvasive imaging with good histopathology correlation.
Collapse
Affiliation(s)
- Pavan P. Adiseshaiah
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Nimit L. Patel
- Small Animal Imaging Program, Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Lilia V. Ileva
- Small Animal Imaging Program, Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Joseph D. Kalen
- Small Animal Imaging Program, Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Diana C. Haines
- Pathology/Histotechnology Laboratory, Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Scott E. McNeil
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| |
Collapse
|
20
|
Using 5-deoxy-5-[18F]fluororibose to glycosylate peptides for positron emission tomography. Nat Protoc 2013; 9:138-45. [PMID: 24356772 DOI: 10.1038/nprot.2013.170] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
So far seven peptide-based (18)F-radiopharmaceuticals for diagnostic applications with positron emission tomography (PET) have entered into clinical trials. Three candidates out of these seven are glycosylated peptides, which may be explained by the beneficial influence of glycosylation on in vivo pharmacokinetics of peptide tracers. This protocol describes the method for labeling peptides with 5-deoxy-5-[(18)F]fluororibose ([(18)F]FDR) as a prosthetic group. The synthesis of [(18)F]FDR is effected by a nucleophilic fluorination step by using dried Kryptofix 2.2.2-K2CO3-K(18)F complex and a subsequent HCl-catalyzed hydrolysis. The conjugation of [(18)F]FDR to the N-terminus aminooxy (-ONH2)-functionalized peptides is carried out in anilinium buffer at pH 4.6 and at room temperature (RT, 21-23 °C), with the concentration of peptide precursors being 0.3 mM. The procedure takes about 120 min and includes two cartridge isolation steps and two reversed-phase (RP) HPLC purification steps. The quaternary methyl amine (QMA) anion exchange cartridge and the hydrophilic-lipophilic balanced (HLB) cartridge are used for the isolation of (18)F-fluoride and [(18)F]FDR-conjugated peptides, respectively. The first HPLC purification provides the (18)F-fluorinated precursor of [(18)F]FDR and the second HPLC purification is to separate labeled peptides from their unlabeled precursors. The final product is formulated in PBS ready for injection, with a radiochemical purity of >98% and a radiochemical yield (RCY) of 27-37% starting from the end of bombardment (EOB). The carbohydrate nature of [(18)F]FDR and the operational convenience of this protocol should facilitate its general use.
Collapse
|