Gallium-68-labeled DOTA-rhenium-cyclized α-melanocyte-stimulating hormone analog for imaging of malignant melanoma

https://doi.org/10.1016/j.nucmedbio.2007.07.003Get rights and content

Abstract

Introduction

Diagnosis of malignant melanoma is critical, since a patient's prognosis is poor. Previous studies have shown that 64Cu- and 86Y-DOTA-ReCCMSH(Arg11) have the potential for early detection of malignant melanoma by exploiting the sensitivity and high resolution of positron emission tomography (PET). This encouraged us to investigate DOTA-ReCCMSH(Arg11) labeled with another β+-emitting radionuclide, 68Ga.

Methods

DOTA-ReCCMSH(Arg11) was successfully labeled with 68Ga at pH 3.8–4 at 85°C. Acute biodistribution and small-animal PET imaging studies were performed in mice bearing B16/F1 melanoma tumor.

Results

Biodistribution studies showed moderate receptor-mediated tumor uptake, fast nontarget organ clearance and high tumor to nontarget tissue ratios. Preadministration of d-lysine significantly reduced kidney uptake without affecting the uptake of the agent in the tumor. Small-animal PET images showed that the tumor could be clearly visualized at all time points examined (0.5–2 h) with the standardized uptake value analysis following a similar trend as the biodistribution data.

Conclusions

The preliminary data obtained suggest that 68Ga-DOTA-ReCCMSH(Arg11) is a promising PET imaging agent for early detection of malignant melanoma.

Introduction

Malignant melanoma, the most serious form of skin cancer, has become a severe health problem due to an increase in incidence and mortality rate [1], [2]. Accurate diagnosis is critical, because unless the primary tumor is excised through surgery, patient prognosis is generally poor [3]. 2-[18F]fluoro-2-deoxy-d-glucose ([18F]FDG) [4], [5], [6] and melanoma-targeting antibodies or antibody fragments [7], [8], [9] have been developed for imaging and staging of disease. [18F]FDG positron emission tomography (PET) is limited in detecting melanoma tumors with small foci [5], and some melanoma cells are undetectable with [18F]FDG because they use non-glucose-based substrates as an energy source [6]; therefore, their clinical application has been limited. Radiolabeled antibodies generally lack specificity due to the presence of individual tumor variants, and since melanomas tend to become amelanotic as the malignancy progresses, the target antigen is often lost due to the reduction in expression of the specific pigment genes that encode this target [10], [11], [12]. The increasing numbers of amelanotic cells in primary tumors or their metastasis increases the possibility that such cells will escape targeting by antibody-based radiopharmaceuticals. Examples of other agents that have been examined clinically for the imaging of melanoma, as alternatives to [18F]FDG, include the PET agents [18F]galacto-RGD [13], 6-[18F]fluoro-l-dopa ([18F]DOPA) [6] and 3′-[18F]fluoro-3′-deoxy-l-thymidine ([18F]FLT) [14] and the SPECT agents N-isopropyl-p-123I-iodoamphetamine ([123I]IMP) [15], 123I-N-(2-diethylaminoethyl)-2-iodobenzamide ([123I]BZA(2)) [16] and 123I-N-[3-(4-morpholino)propyl]-N-methyl-2-hydroxy-5-iodo-3-methylbenzylamine (ERC9) [17].

Recently, alpha-melanocyte-stimulating hormone (α-MSH) peptide analogs were developed as promising agents for both melanoma imaging and radiotherapy. α-MSH is a tridecapeptide that is excreted by the pars intermedia of the pituitary gland and is primarily responsible for the regulation of skin pigmentation [18], [19]. α-MSH targets the melanocortin-1 receptor (MC1R), which belongs to a family of G-protein-coupled receptors (MC1R to MC5R) that have been isolated in both mice and humans [20], [21], [22], [23], [24], [25], [26]. These receptors are normally expressed on the surface of melanocytes, and it was reported that >80% of human melanoma tumor samples obtained from patients with metastatic lesions express α-MSH receptors [27], [28]. It has been reported the MC1R expression ranges from several hundred to around 10,000 receptors per cell [29]. In 2003, Miao et al. [30] reported that MC1R receptor expression ranged from 900 (1.49 fmol per million SKMEL28 cells) to around 6000 (9.46 fmol per million TXM13 cells) receptors per cell on human melanoma cells, enabling the use of radiolabeled α-MSH peptide analogs as specific melanoma diagnostic and therapeutic agents.

There have been a number of reports on α-MSH peptide-based agents that have demonstrated the ability to image the MCR1 receptor in vivo [31], [32], [33], [34], [35], [36], [37], [38], [39]. We previously described an α-MSH-targeting peptide system, DOTA-ReCCMSH(Arg11), which was labeled with β+-emitting radiometals, as potential agents for the detection of malignant melanoma via PET imaging [40]. The rhenium-cyclized peptide system was chosen for further study given its increased stability compared to its linear analog with a concomitant increase in tumor uptake and less renal retention [38], [39]. We chose the divalent metal ion 64Cu (t1/2=12.7 h, 17.4% β+, 41% electron capture (EC), 40% β) [41] and trivalent metal ion 86Y (t1/2=14.7 h, 33% β+), both of which can be prepared on a biomedical cyclotron utilizing the 64Ni(p,n)64Cu and 86Sr(p,n)86Y reactions, respectively [42], [43]. Our results showed that both 64Cu-DOTA-ReCCMSH(Arg11) and 86Y-DOTA-ReCCMSH(Arg11) have the potential for early detection of malignant melanoma by exploiting the sensitivity and high resolution of PET [40]. This encouraging result motivated us to investigate the same peptide DOTA-ReCCMSH(Arg11) radiolabeled with another β+-emitting radiometal 68Ga. 68Ga (t1/2=68 min, 88% β+) is a short-lived positron emitter, the production of which is not dependent on an in-house cyclotron since it is available from commercially available 68Ge/68Ga generators that have >1-year shelf lives. The use of 68Ga-labeled peptides is an emerging area of PET radiopharmaceutical development and a number of agents have been translated to the clinic [44]. This report describes our preliminary study on 68Ga labeled DOTA-ReCCMSH(Arg11), and, to the best of our knowledge, this is the first study on 68Ga-labeled DOTA-conjugated cyclized α-MSH peptide analog. We report small-animal PET and biodistribution studies of 68Ga-DOTA-ReCCMSH(Arg11) in the B16/F1 tumor-bearing mouse model.

Section snippets

General

Unless otherwise stated, all chemicals were purchased from Sigma Aldrich Chemical Co. (St. Louis, MO, USA). Water was distilled and then deionized (18 MΩ/cm2) by passing through a Milli-Q water filtration system (Millipore Corp., Milford, MA, USA). 68Ga was obtained from a 68Ge/68Ga radionuclide generator (TCI Medical Inc., Albuquerque, MN, USA; 30 mCi generator at delivery). The parent, 68Ge, is accelerator produced and decays with a half-life of 270.8 days by electron capture. This 68Ge

68Ga labeling of DOTA-ReCCMSH(Arg11)

DOTA-ReCCMSH(Arg11) was successfully labeled with 68Ga at 85°C. It was found that an equal volume of 1.25 M ammonium acetate (pH 4.5), added to 68GaCl3 in 0.1 M HCl, resulted in a final pH of 3.8–4. The pH range was found to be critical for the successful labeling of DOTA-ReCCMSH(Arg11) with 68Ga. The radiolabeling yield was much lower (<10%) if the final pH of the reaction mixture was 3.2–3.5 or when 0.1 or 0.5 M ammonium acetate (pH 4.5) was used instead of 1.25 M ammonium acetate. Using the

Discussion

Several radiolabeled α-MSH peptides have been investigated for melanoma-specific targeting. Substitution of Met4 with Nle4 and Phe7 with d-Phe7 yielded the [NIe4 D-Phe7]α-MSH (NDP) analogs that showed subnanomolar receptor affinity and resistance to enzymatic degradation [29], [47], [48]. α-MSH peptides cyclized via a disulfide bond [Cys(4,10), d-Phe7]α-MSH [49] and lactam bond formation [Asp5, d-Phe7, Lys11 α-MSH] [50], [51] display increased receptor-binding affinity and resistance to

Conclusions

68Ga-DOTA-ReCCMSH(Arg11) was evaluated for the PET imaging of malignant melanoma in the B16/F1 tumor-bearing mouse model. The high tumor to nontarget tissue ratios, fast clearance from nontarget organs and moderate tumor uptake suggest that 68Ga labeled DOTA-ReCCMSH(Arg11) is a promising agent for the detection of malignant melanoma. The tumor uptake of 68Ga-DOTA-ReCCMSH(Arg11) is not optimal due to its low effective specific activity.

Acknowledgments

We are very grateful for the technical assistance of Dawn Werner, Terry Sharp, Lori Strong, Nicole Fettig, Margaret Morris, Amanda Roth, Ann Stroncek and Jerrel Rutlin. We further wish to thank Dr. Raffaella Rossin for her help and assistance with the gallium generator. This work was partially supported by the National Cancer Institute (NCI) (R24 CA86307 and P50 CA103130). Small-animal PET imaging is supported by a National Institutes of Health/NCI SAIRP grant (R24 CA86060) with additional

References (66)

  • F. Vosmik

    Malignant melanoma of the skin. Epidemiology, risk factors, clinical diagnosis

    Csa Lek Cesk

    (1996)
  • A.N. Eberle et al.

    Radiolabeled alpha-melanocyte-stimulating hormone analogs for receptor-mediated targeting of melanoma: from tritium to indium

    J Mol Recognit

    (2003)
  • P. Som et al.

    A fluorinated glucose analog, 2-fluoro-2-deoxy-d-glucose (F-18): nontoxic tracer for rapid tumor detection

    J Nucl Med

    (1980)
  • G.S. Mijnhout et al.

    Systematic review of the diagnostic accuracy of 18F-fluorodeoxyglucose positron emission tomography in melanoma patients

    Cancer

    (2001)
  • A. Dimitrakopoulou-Strauss et al.

    Quantitative PET studies in pretreated melanoma patients: a comparison of 6-[18F]fluoro-l-DOPA with 18F-FDG and 15O-water using compartment and noncompartment analysis

    J Nucl Med

    (2001)
  • D. Salk

    Technetium-labeled monoclonal antibodies for imaging metastatic melanoma: results of a multicenter clinical study

    Semin Oncol

    (1988)
  • A. Taylor et al.

    Radioimmunodetection of human melanoma with indium-111-labeled monoclonal antibody

    J Nucl Med

    (1988)
  • K.S. Marchitto et al.

    Characterization of a human–mouse chimeric antibody reactive with a human melanoma associated antigen

    Prog Clin Biol Res

    (1989)
  • M.J. Adler et al.

    Amelanotic malignant melanoma

    Semin Cutan Med Surg

    (1997)
  • T.G. Eisen

    The control of gene expression in melanocytes and melanomas

    Melanoma Res

    (1996)
  • V.J. McGovern

    Melanoma: histological diagnosis and prognosis

    (1983)
  • A.J. Beer et al.

    Biodistribution and pharmacokinetics of the alphavbeta3-selective tracer 18F-galacto-RGD in cancer patients

    J Nucl Med

    (2005)
  • D.C. Cobben et al.

    3′-18F-Fluoro-3′-deoxy-l-thymidine: a new tracer for staging metastatic melanoma?

    J Nucl Med

    (2003)
  • K. Kato et al.

    Low efficacy of 18F-FDG PET for detection of uveal malignant melanoma compared with 123I-IMP SPECT

    J Nucl Med

    (2006)
  • N. Moins et al.

    123I-N-(2-diethylaminoethyl)-2-iodobenzamide: a potential imaging agent for cutaneous melanoma staging

    Eur J Nucl Med Mol Imaging

    (2002)
  • T.G. Salopek et al.

    Radioiodinated N-[3-(4-morpholino)propyl]-N-methyl-2-hydroxy-5-iodo-3-methylbenzylamine (ERC9): a new potential melanoma imaging agent

    Eur J Nucl Med

    (2001)
  • R.D. Cone et al.

    The melanocortin receptors: agonists, antagonists, and the hormonal control of pigmentation

    Recent Prog Horm Res

    (1996)
  • R.D. Cone et al.

    Cloning and functional characterization of a family of receptors for the melanotropic peptides

    Ann N Y Acad Sci

    (1993)
  • P. Barrett et al.

    Cloning and expression of a new member of the melanocyte-stimulating hormone receptor family

    J Mol Endocrinol

    (1994)
  • F. Desarnaud et al.

    Molecular cloning, functional expression and pharmacological characterization of a mouse melanocortin receptor gene

    Biochem J

    (1994)
  • O. Labbe et al.

    Molecular cloning of a mouse melanocortin 5 receptor gene widely expressed in peripheral tissues

    Biochemistry

    (1994)
  • A.N. Eberle
  • J.B. Tatro et al.

    Interaction of an alpha-melanocyte-stimulating hormone-diphtheria toxin fusion protein with melanotropin receptors in human melanoma metastases

    Cancer Res

    (1992)
  • Cited by (51)

    • Biomedical applications of radioiodinated peptides

      2019, European Journal of Medicinal Chemistry
      Citation Excerpt :

      and references therein The overexpression of MC1R in more than 80% of human metastatic melanoma turned it into a potential relevant target for imaging and systemic radiotherapy [145]. In recent years, several radiolabelled α-MSH analogues have been explored as potential candidates for MC1R targeting and have been evaluated for both melanoma imaging and radionuclide therapy [147–151]. SAR studies have identified the amino acid sequence His-Phe-Arg-Trp as the minimal motif required for biological activity.

    • Optimizing the radiosynthesis of [<sup>68</sup>Ga]DOTA-MLN6907 peptide containing three disulfide cyclization bonds – a GCC specific chelate for clinical radiopharmaceuticals

      2018, Applied Radiation and Isotopes
      Citation Excerpt :

      Radiochemical purity and efficiency were determined by instant thin layer chromatography (iTLC) and high performance liquid chromatography (HPLC); combined with radioactivity detection (radioHPLC). Radio-iTLC was conducted using a Bioscan AR-200 radio-chromatographic scanner to scan radioactivity distributions on iTLC-SG plates (Varian, Lake Forest, CA 92630; Cat# SGI0001) by methods described previously (Jeong et al., 2008; Wei et al., 2007). Briefly, iTLC material was cut into 10 cm x 1.5 cm strips and baked at 100 °C for at least 3 h and allowed to stand no more than one hour prior to use.

    • Optimization of the solid-phase synthesis of [<sup>18</sup>F] radiolabeled peptides for positron emission tomography

      2012, Applied Radiation and Isotopes
      Citation Excerpt :

      Peptides play an increasingly important role as in vivo molecular imaging agents for the early, non-invasive detection and localization of many diseases including cancer (Bergmann et al., 2002; Ginj et al., 2006; Hoffman et al., 2003; Lee et al., 2010; Wei et al., 2007).

    View all citing articles on Scopus
    View full text