Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 May 1.
Published in final edited form as: Curr Opin Oncol. 2010 May;22(3):274–280. doi: 10.1097/CCO.0b013e3283373d5c

“Image and Treat” – An Individualized Approach to Urological Tumors

Kirsten Bouchelouche 1, Jacek Capala 2
PMCID: PMC2855391  NIHMSID: NIHMS189963  PMID: 20150811

Abstract

Purpose of review

The current treatment options for advanced urologic cancers demonstrate limited efficacy. To obtain optimal clinical results, there is a need for new, individualized, therapeutic strategies, which has only recently been applied to these malignancies. Nuclear medicine plays an increasing role in establishing imaging biomarkers necessary for personalized medicine. This review focuses on the current status of the “image and treat” approach combining molecular imaging with targeted radionuclide therapy of urological malignancies

Recent findings

Tumour-specific targets in uro-oncology are showing promising results for development of personalized therapy using positron emission tomography/computed tomography (PET/CT) molecular imaging and radioimmunotherapy (RIT). The antibody cG250, which binds to carbonic anhydrase IX (CAIX), is being evaluated as a radiolabelled imaging and therapeutic agent in clear cell renal cell carcinoma (ccRCC). 124I -cG250 PET/CT has demonstrated excellent targeting of ccRCC. Prostate specific membrane antigen (PSMA) is a promising target for both PET/CT and RIT of prostate cancer. HER2 may be another potential target in bladder and prostate cancer.

Summary

Tumour-specific targets and biomarkers are being studied for PET/CT and RIT. This may lead to development of new therapeutic strategies. However, considerable investment in new research will be required for personalized medicine to be routinely used in uro-oncology.

Keywords: positron-emission tomography, radioimmunotherapy, PSMA, CAIX, G250, HER2 receptor

Introduction

Tumour resistance and toxicity to normal tissues limit the efficacy of conventional anti-cancer treatments such as radiation and chemotherapy. The systemic side effects of chemotherapy in particular represent a severe problem. The most promising alternative to conventional therapy is targeted tumor therapy where selective molecules are used to direct anti-cancer drugs to the tumor, thereby limiting the damage to healthy tissue. For targeted tumor therapy, peptides and antibodies possess a key position as drug delivery vectors [1]. To kill the targeted tumour cells, the favourable targeting properties of antibodies and peptides must be combined with an efficient cytotoxic moiety such as toxins, drugs, or therapeutic radioactive nuclides. Targeted radionuclide therapy (TRT) uses the latter as a local source of radiation to combine the favourable targeting properties of peptides and antibodies with the effectiveness of radiation-induced cell death [2;3]. A major advantage of TRT is the possibility to determine the selective accumulation in the targeted tissue by molecular imaging studies via single photon computed tomography (SPECT) or positron emission tomography (PET) using structurally identical diagnostic compounds. For this purpose, targeting of epitopes that are expressed even in relatively low concentrations is feasible. These non-invasive imaging methods allow estimation of radiation dose distribution prior to therapy, tumour staging, and early monitoring of the efficacy of individual treatments. This novel class of pharmaceuticals offers the potential to develop patient-specific therapies based on the new “image and treat” approach.

The current treatment options for advanced urologic cancers demonstrate limited efficacy and severe side effects [••4,•5,••6,7]. Therefore, there is a need for new therapeutic strategies. The idea of individualizing therapies to obtain optimal clinical results is not new but has only recently been applied to urological malignancies. This review focuses on the recent advances of molecular imaging as applied to TRT in uro-oncology.

Molecular imaging and targeted radionuclide therapy

In recent years there has been much focus on personalized medicine, where pharmaceutical therapies are tailored to the particular characteristics of the individual patient. The role of molecular imaging in personalized medicine, using targeted drugs in oncology, is very attractive. It provides in vivo regional information before and, in real time, during treatment, which cannot be obtained by in vitro methods (“regional proteomics”). In clinical practice, imaging biomarkers may be used to screen for cancer, confirm diagnosis, assess extent, and predict response to available therapies [8]. Nuclear medicine plays an important role in establishing imaging biomarkers in clinical decision-making. 18F-fluorodeoxyglucose (FDG), the first PET molecular imaging biomarker, is a biomarker of glucose metabolism. Most cancer cell types demonstrate increased glucose metabolism leading to increased 18F-FDG uptake. 18F–FDG PET/CT is now widely used in the management of several cancer types. In uro-oncology, PET/CT has been one of the slowest areas to develop. This is mainly due to urinary excretion of 18F–FDG and low 18F–FDG uptake especially in prostate and some renal cancers. However, the role of PET/CT in uro-oncology is likely to expand as new and more favourable tracers are evaluated [9]. New biomarkers that image cell proliferation, apoptosis, angiogenesis, hypoxia, and growth factor receptors are being studied [10], and may lead to enhanced clinical management of cancer patients.

Radiolabelling of modified forms of the imaging biomolecules with therapeutic radionuclides allows therapeutic applications using radioconjugates that are almost identical to the imaging probes. In contrast to chemotherapy, TRT requires very low mass amounts of the targeting compound. Peptide receptor radiation therapy (PRRT), as one form of TRT, is based on site-specific accumulation, preferentially due to receptor-mediated endocytosis and intracellular retention of radiolabelled peptides [10;11]. At present, PRRT is mainly used for the treatment of neuroendocrine tumours [12]. The somatostatin receptor-binding agents 90Y-DOTATOC and 177Lu-DOTATATE are examples of successfully applied oncological PRRTs [2;3;13]. In radioimmunotherapy (RIT), antibodies are labelled with radionuclides for therapy. 90Y-rituximab (Zevalin®, Biogen, IDEC, Cambridge, MA) and 131I-tositumomab (Bexxar®, Corixa, Seattle, WA, USA) are examples of radiolabelled antibodies for cancer treatment [••13]. These two therapeutic agents target the CD20-antigen on B-cells.

The therapeutic effect of TRT is achieved primarily by ionization radiation of the radionuclide, and the therapeutically effective radiation dose is determined by the physical characteristics of the radionuclide. As the radiation is not restricted to the targeted cell, it may also affect all tumor cells in its range. This effect, called “bystander” or “crossfire” effect, is particularly important for treatment of tumors with heterogeneous antigen or receptor expression or insufficient vascularization [2]. The most common nuclides that are currently used for endoradiotherapy are β-emitters such as 131I, 177Lu, and 90Y. Medium-energy β-emitters, i.e., 131I and 177Lu, are more effective for the treatment of small tumors [2;3]. In larger tumors, isotopes emitting high-energy β-radiation, like 90Y, might present a better alternative. Molecular imaging plays an essential role in balancing the clinical benefits and risks of TRT. To effectively treat individual patients, careful assessment of biodistribution, dosimetry, and toxicity of the therapeutic radionuclide is essential.

Carbonic anhydrase-IX

Renal cell carcinoma (RCC) is the third most common genitourinary cancer site after prostate and bladder cancer [•14]. Imaging plays an important role in the clinical management of RCC [••15]. However, RCC is a radio- and -chemotherapy resistant tumor, and current treatments have limited efficacy, resulting in high morbidity and mortality in patients with metastatic disease [4]. Thus, there is a need for new therapeutic strategies in RCC. Monoclonal antibodies targeting tumor-associated antigens have been developed for RCC [4;16], and targeted agents are being used increasingly for the treatment of metastatic RCC [17;18]. Among them, targeting of carbonic anhydrase IX (CAIX) antigen using monoclonal chimeric G250 antibody (cG250) is a new approach for imaging and treating RCC. CAIX is ubiquitously expressed in more than 90% of clear-cell RCC (ccRCC) but not in normal kidney [16;1921]. In addition, high expression of CAIX appears to be a marker of poor prognosis in ccRCC [20;22].

cG250 PET

The antibody G250 has been studied in ccRCC both as a murine antibody and a chimeric antibody (cG250) [23]. For molecular imaging, radiolabelled cG250 demonstrates excellent visualization of known metastatic lesions, and frequently also detects new lesions in patients [2428]. Recently, 124I-cG250 has been used for evaluating RCC. Divgi et al. assessed whether 124I-cG250 PET could predict for ccRCC in 26 patients with renal masses [28]. Surgery was scheduled one week after 124I-cG250 infusion. PET/CT scanning was performed within 3 h before surgery. In this series, 124I-cG250 PET accurately identified 15 of 16 ccRCC, and all nine non-clear-cell renal masses were negative for the tracer. The sensitivity of 124I-cG250 PET was 94%, the negative predictive value was 90%, and specificity and positive predictive accuracy were both 100%. Currently, a multicenter trial in a larger group of patients is being conducted to evaluate the role of 124I-cG250 PET/CT in patients with renal masses [23]. 124I-cG250 PET/CT may be a valuable tool in diagnosing metastases in patients with a G250 positive primary tumor and/or in the differential diagnosis of suspect kidney lesions, i.e., distinguishing ccRCC from other subtypes. Preoperative identification of tumor type could have important implications for the choice of treatment.

CAIX is associated with hypoxia, and expression of CAIX is regulated by the hypoxia-inducible factor 1α (HIF-1α) [29]. Recently, Lawrentschuk et al. used in-vivo studies for investigation of hypoxia and CAIX expression in a RCC xenograft model with oxygen tension measurements and 124I-cG250 PET/CT [••30]. 124I-cG250 PET/CT demonstrated excellent tumor targeting, and a correlation between tracer uptake as measured by standard uptake value (SUV) on non-invasive PET/CT studies and traditional biodistribution studies was demonstrated. However, no significant correlation between CAIX and hypoxia was found.

cG250 radioimmunotherapy

The first G250 investigated for RIT in ccRCC were murine antibodies, which can cause an immune response in humans. The production of human-anti-mouse-antibodies (HAMA) inhibits the effectiveness of the second administrated dose of radiolabelled antibody [3]. In a Phase I/II RIT study, escalating activity doses of 131I-G250 were administrated to patients with metastatic RCC [31]. Seventeen out of 33 patients showed stabilization of disease progression. However, all the patients developed a HAMA response. After development of the chimeric form of G250, 131I-cG250 was tested in clinical trials [25;32;33]. The maximum tolerated dose (MTD) of 131I-cG250 was determined in a phase I trial in patients with advanced metastatic RCC [24]. The MTD was determined to be 2220 MBq/m2, with hematological toxicity being the dose-limiting factor. Divgi et al. performed fractionation of the dose in a Phase I study [33]. Steffens et al. investigated the effect of two sequential high doses of 131I-cG250 [25]. Five of the 18 patients evaluated had stabilization of their disease, lasting 3–12 months. However, no partial or complete responses were seen.

Radionuclides other than 131I may enhance the therapeutic index of radiolabelled cG250. In order to optimize cG250 RIT, the therapeutic properties of cG250 labeled with four different radionuclides were tested in mice with ccRCC xenografts [34]. The results of the in vivo study indicated that 177Lu- and 90Y-cG250 conjugates may be superior for RIT compared to 131I conjugates. Currently, a phase I/II trial is ongoing, investigating the efficacy of 177Lu-DOTA-cG250 in patients with advanced RCC (NCT00142415). Preliminary results demonstrate excellent tumor targeting of RCC lesions and indicate that 177Lu-cG250 treatment can stabilize previously progressive metastatic RCC [35].

Prostate specific membrane antigen

Prostate cancer is the most common cancer in US men, and is the second leading cause of cancer death in men in the United States [•36]. Imaging is important in the clinical management of prostate cancer patients [••37]. Treatments range from surveillance to radical local treatment to androgen-deprivation therapy [7]. Since there are no effective treatments for advanced prostate cancer, new strategies have to be considered [36]. Several cell surface proteins, glycoproteins, receptors, enzymes, and peptides have been proposed as potential targets for the treatment of prostate cancer [38;39]. Among these, the prostate specific membrane antigen (PSMA) represents an attractive target for molecular imaging and therapy [•40]. PSMA is specifically expressed on prostate epithelial cells, and is strongly up regulated in prostate cancer, with highest expression occurring in androgen insensitive or metastatic disease [•41]. PSMA is not secreted or released into the circulation unlike prostate specific antigen (PSA), which makes it an excellent target for diagnostic and therapeutic agents.

PSMA imaging

The first commercial monoclonal antibody (mAb) against PSMA was 7E11, which is used in the FDA-approved 111In-CYT-356-based imaging of prostate cancer (ProstaScint) [42]. The 7E11 antibody targets an intracellular epitope of PSMA, and therefore binds only to permeabilized necrotic cells [41]. Another anti-PSMA mAb, J591, which binds to an epitope on the extracellular domain of PSMA [43], presents a better alternative for targeting of prostate cancer. J591 had been extensively studied in preclinical models, and demonstrated high tumor-to-normal tissue ratios in prostate cancer xenografts [44;45]. These studies were followed by several clinical trials using J591 labelled with different nuclides for radioimmunoscintigraphy and RIT [4648] that confirmed the feasibility of J591 as a prostate cancer-targeting agent (see PSMA radioimmunotherapy below).

Recently, other mAbs that target PSMA for molecular imaging have been developed. Elsässer-Beile et al., reported the development of three IgG mAbs (3/A12, 3/E7, and 3/F11) with affinity for PSMA [49]. Wolf et al., demonstrated that 3/A12, 3/E7, and 3/F11 bind to different extracellular epitopes of PSMA [•50]. Elsässer+-Beile et al, used 64Cu-3/A12 for PET imaging in a prostate cancer xenograft model [••51]. PET was performed 3, 24, and 48 h after injection of 64Cu-3/A12, and good tumor-to-background ratio was found. PSMA-negative tumors were negative on PET. Low-molecular-weight, radiopharmaceutical-based imaging agents may provide superior pharmacokinetics for imaging than radiolabelled antibodies characterized by long circulation time and delayed clearance from non-target tissues [52]. Recently, low-molecular-weight agents that target or inhibit PSMA have demonstrated promising results [53;54;5456, ••57]. Foss et al., showed successful imaging of xenografts that express PSMA using PET and the radiolabelled PSMA inhibitor N-[N-[(S)-1,3-dicarboxypropyl]carbamoyl]-S-[11C]methyl-l-cysteine (DCFBC) [54]. Mease et al., extended that work by preparing and testing a PSMA inhibitor of the same class labelled with 18F [53]. Biodistribution and imaging studies showed high uptake of 18F-DCFBC in the PSMA positive tumors with little to no uptake in PSMA negative tumors. Hillier et al., reported that two small molecule inhibitors targeting PSMA, MIP-1072 and MIP-1095, exhibited high affinity for PSMA [••57]. The uptake of 123I-MIP-1072 and 123I-MIP-1095 in prostate cancer xenografts was shown by SPECT/CT.

PSMA radioimmunotherapy

Prostate cancer represents an attractive target for RIT for several reasons: 1) the prostate gland is a nonvital organ, thereby allowing targeting of tissue-specific antigens, 2) metastases from prostate cancer are mainly localized in lymph nodes and bones, locations with good access to circulating antibodies, 3) the metastases are often small enough to ensure good antibody penetration, and 4) the serum marker PSA can be used for monitoring of therapeutic efficacy [4042;58]. In RIT, myelotoxicity due to bone marrow radiation-absorbed doses (Bmrad) is frequently the dose-limiting factor that determines the maximum tolerated dose (MTD). In a dose-escalation study, prostate cancer patients (n=28) were treated with either 90Y- or 177Lu-labelled J591 antibodies [59]. Myelotoxicity after treatment with 177Lu-J591 could be predicted on the basis of the radioactive dose administered or the Bmrad. In contrast, no correlation between myelotoxicity and 90Y-J591 dose was found. In a phase I trial, the MTD of 177Lu-J591 was found to be 70 mCi/m2 in advanced prostate cancer patients [60]. Multiple doses of 30 mCi/m2 were well tolerated, and excellent targeting of known metastases was demonstrated.

In a recent study by Pandit-Tascar et al., 14 patients with metastatic prostate cancer received escalating doses of 111In-J591 in a series of administrations, each separated by 3 weeks [48]. 111In-J591 correctly localized 93.7% of the bone lesions detected by conventional imaging. In the study, the optimal antibody mass for RIT was determined to be greater than or equal to 50 mg. Recently, Tagawa et al. showed in a phase II trial that a single dose of 177Lu-J591 was well tolerated, and with reversible myelosuppression. In the study, anti-tumor activity was demonstrated in patients with advanced metastatic castrate-resistant prostate cancer, and excellent targeting of known sites of metastases was seen in 97% (31/32) of the patients [47].

HER2

Overexpression of the HER2 protein and amplification of the HER2 gene have been implicated in tumor development, progression, and poor prognosis in several types of cancers [61]. There is increasing evidence that HER2 also plays a role in advanced prostate cancer [6264] and bladder cancer [65;66]. In prostate cancer patients, HER2 expression is associated with disease progression and androgen independence [62;63], and preoperative plasma HER2 is associated with cancer progression after prostatectomy [62;64]. Caner et al., found that 61.1 % of high-grade urothelial carcinomas demonstrated HER2 overexpression [67]. Recently, Laé et al demonstrated in a large study (n=1005) that 9.2 % of invasive bladder carcinomas are HER2 positive [66]. Therefore, HER2 represents a potential target for both molecular imaging and therapy in HER2 positive bladder and prostate cancers.

HER2 imaging

Monoclonal antibodies such as trastuzumab and pertuzumab, or small scaffold Affibody molecules, are used as HER2-targeting agents [61]. For imaging purposes, these agents are labelled with positron- or gamma-emitting radionuclides for PET or SPECT imaging, respectively [61; ••68,69,7073]. There is increasing evidence that Affibody molecules or other small non-immunoglobulin based tracers have the best potential for developing high-contrast imaging agents to visualize HER2 in vivo compared to full-length monoclonal antibodies [61;72]. Kramer-Marek et al. have radiolabelled a HER2-binding Affibody molecule with 18F for in vivo monitoring of HER2 expression by PET [73]. Recently, Kramer-Marek et al. also demonstrated that the same tracer can be used for PET/CT imaging to assess changes in HER2 expression following therapeutic intervention [••74]. Baum et al. used HER2 Affibody molecules labelled with 111In or 68Ga, and demonstrated high quality SPECT and PET/CT imaging of HER2 positive xenografts [75]. It was possible to detect even very small malignant lesions. Cheng et al. also found PET imaging of HER2 expression promising in a xenograft tumor model [71]. Currently, an ongoing trial is evaluating the role of 111In-CHX-A DTPA Trastuzumab imaging for HER2 expression in breast cancer patients (NCI-07-C-0101).

HER2 radionuclide therapy

For therapy, the targeting vector (trastuzumab, Affibody molecules) has been labelled with radionuclides suitable for therapy [72;76, •77,7879]. Tolmachev et al. labelled a HER2-specific Affibody molecule with 177Lu for radionuclide therapy of HER2-positive microxenografts. The results indicate that HER2 RIT may be promising for the treatment of HER2-expressing malignant micrometastases [76]. This strategy, involving assessment of target presence (HER2 positive cancer) and distribution in an individual patient, followed by optimized HER2-specific radionuclide drug delivery, has the potential to improve therapeutic outcome of HER2 positive cancers while reducing side effects.

CONCLUSION

The aim of today’s state of the art molecular imaging is detection of the very early stages of cancer, followed by therapeutic action. Molecular imaging may provide unique means for the selection of patients who may benefit from targeted therapies, as well as allow monitoring of early responses to treatment, and enable subsequent restaging. A considerable amount of effort has been focused on the development of personalized medicine in oncology. In recent years, tumour-specific biomarkers have proven to be potentially useful in the development of new therapeutic strategies in uro-oncology. However, considerable investments in research are still required for personalized medicine to be fully developed and implemented clinically.

Abbreviations

BMrad

bone marrow radiation-absorbed doses

CAIX

carbonic anhydrase IX

ccRCC

clear cell renal cell carcinoma

CT

computerized tomography

FDG

fluorodeoxyglucose

HAMA

human-anti-mouse-antibodies

mAb

monoclonal antibody

MTD

maximum tolerated dose

PET

positron emission tomography

PPRT

peptide receptor radiation therapy

PSMA

prostate specific membrane antigen

RCC

renal cell carcinoma

RIT

radioimmunotherapy

SPECT

single photon computed tomography

TRT

targeted radionuclide therapy

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Reference List

  • 1.Brissette R, Prendergast JK, Goldstein NI. Identification of cancer targets and therapeutics using phage display. Curr Opin Drug Discov.Devel. 2006;9:363–369. [PubMed] [Google Scholar]
  • 2.Wangler C, Buchmann I, Eisenhut M, Haberkorn U, Mier W. Radiolabeled peptides and proteins in cancer therapy. Protein Pept.Lett. 2007;14:273–279. doi: 10.2174/092986607780090874. [DOI] [PubMed] [Google Scholar]
  • 3.Boerman OC, Koppe MJ, Postema EJ, Corstens FH, Oyen WJ. Radionuclide therapy of cancer with radiolabeled antibodies. Anticancer Agents Med Chem. 2007;7:335–343. doi: 10.2174/187152007780618126. [DOI] [PubMed] [Google Scholar]
  • 4. Rini BI, Campbell SC, Escudier B. Renal cell carcinoma. Lancet. 2009;373:1119–1132. doi: 10.1016/S0140-6736(09)60229-4. •• Excellent review on epidemiology, diagnosis, surgical and systemic treatment strategies of RCC.
  • 5. Bischoff CJ, Clark PE. Bladder cancer. Curr Opin Oncol. 2009;21:272–277. doi: 10.1097/cco.0b013e328329f184. • The authors review briefly the diagnosis and management of all stages of bladder cancer, with emphasis on recent developments in 2008
  • 6. Kaufman DS, Shipley WU, Feldman AS. Bladder cancer. Lancet. 2009;374:239–249. doi: 10.1016/S0140-6736(09)60491-8. •• Also a very good review on bladder cancer with updated information on presentation, diagnosis and treatment strategies.
  • 7.Damber JE, Aus G. Prostate cancer. Lancet. 2008;371:1710–1721. doi: 10.1016/S0140-6736(08)60729-1. [DOI] [PubMed] [Google Scholar]
  • 8.Carrio I. Key role of nuclear medicine in bringing imaging biomarkers into clinical practice. Eur J Nucl Med Mol.Imaging. 2009;36:1937–1939. doi: 10.1007/s00259-009-1318-3. [DOI] [PubMed] [Google Scholar]
  • 9.Bouchelouche K, Oehr P. Positron emission tomography and positron emission tomography/computerized tomography of urological malignancies: an update review. J Urol. 2008;179:34–45. doi: 10.1016/j.juro.2007.08.176. [DOI] [PubMed] [Google Scholar]
  • 10.Oyen WJ, Bodei L, Giammarile F, Maecke HR, Tennvall J, Luster M, Brans B. Targeted therapy in nuclear medicine--current status and future prospects. Ann.Oncol. 2007;18:1782–1792. doi: 10.1093/annonc/mdm111. [DOI] [PubMed] [Google Scholar]
  • 11.Haberkorn U, Eisenhut M, Altmann A, Mier W. Endoradiotherapy with peptides - status and future development. Curr Med Chem. 2008;15:219–234. doi: 10.2174/092986708783497256. [DOI] [PubMed] [Google Scholar]
  • 12.de JM, Breeman WA, Kwekkeboom DJ, Valkema R, Krenning EP. Tumor imaging and therapy using radiolabeled somatostatin analogues. Acc.Chem.Res. 2009;42:873–880. doi: 10.1021/ar800188e. [DOI] [PubMed] [Google Scholar]
  • 13. Zoller F, Eisenhut M, Haberkorn U, Mier W. Endoradiotherapy in cancer treatment--basic concepts and future trends. Eur J Pharmacol. 2009;625:55–62. doi: 10.1016/j.ejphar.2009.05.035. •• The article reviews endoradiotherapy in cancer, including peptide receptor radiation therapy and radioimmunotherapy.
  • 14. Garcia JA, Cowey CL, Godley PA. Renal cell carcinoma. Curr Opin Oncol. 2009;21:266–271. doi: 10.1097/CCO.0b013e32832a05c8. • The authors review recent developments in molecular biomarkers and treatment of advanced RCC
  • 15. Choudhary S, Sudarshan S, Choyke PL, Prasad SR. Renal cell carcinoma: recent advances in genetics and imaging. Semin.Ultrasound CT MR. 2009;30:315–325. doi: 10.1053/j.sult.2009.03.003. •• This review focuses on recent advances in the molecular biology and cytogenetics of RCC, with important diagnostic, therapeutic and prognostic implications
  • 16.Stillebroer AB, Oosterwijk E, Oyen WJ, Mulders PF, Boerman OC. Radiolabeled antibodies in renal cell carcinoma. Cancer Imaging. 2007;7:179–188. doi: 10.1102/1470-7330.2007.0025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Patard JJ, Pouessel D, Bensalah K, Culine S. Targeted therapy in renal cell carcinoma. World J Urol. 2008;26:135–140. doi: 10.1007/s00345-008-0237-4. [DOI] [PubMed] [Google Scholar]
  • 18.Oosterwijk E, Boerman OC, Oyen WJ, Old LJ, Mulders PF. Antibody therapy in renal cell carcinoma. World J Urol. 2008;26:141–146. doi: 10.1007/s00345-008-0236-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Oosterwijk E, Ruiter DJ, Hoedemaeker PJ, Pauwels EK, Jonas U, Zwartendijk J, Warnaar SO. Monoclonal antibody G 250 recognizes a determinant present in renal-cell carcinoma and absent from normal kidney. Int.J Cancer. 1986;38:489–494. doi: 10.1002/ijc.2910380406. [DOI] [PubMed] [Google Scholar]
  • 20.Oosterwijk E. Carbonic anhydrase IX: historical and future perspectives. BJU.Int. 2008;101 Suppl 4:2–7. doi: 10.1111/j.1464-410X.2008.07641.x. [DOI] [PubMed] [Google Scholar]
  • 21.Jensen HK, Nordsmark M, Donskov F, Marcussen N, von der MH. Immunohistochemical expression of carbonic anhydrase IX assessed over time and during treatment in renal cell carcinoma. BJU.Int. 2008;101 Suppl 4:41–44. doi: 10.1111/j.1464-410X.2008.07649.x. [DOI] [PubMed] [Google Scholar]
  • 22.Belldegrun AS, Bevan P. Carbonic anhydrase IX: role in diagnosis, prognosis and cancer therapy. Introduction. BJU.Int. 2008;101 Suppl 4:1. doi: 10.1111/j.1464-410X.2008.07640.x. [DOI] [PubMed] [Google Scholar]
  • 23.Perini R, Pryma D, Divgi C. Molecular imaging of renal cell carcinoma. Urol Clin.North Am. 2008;35:605–611. doi: 10.1016/j.ucl.2008.07.015. [DOI] [PubMed] [Google Scholar]
  • 24.Steffens MG, Boerman OC, de Mulder PH, Oyen WJ, Buijs WC, Witjes JA, van den Broek WJ, Oosterwijk-Wakka JC, Debruyne FM, Corstens FH, Oosterwijk E. Phase I radioimmunotherapy of metastatic renal cell carcinoma with 131I-labeled chimeric monoclonal antibody G250. Clin.Cancer Res. 1999;5:3268s–3274s. [PubMed] [Google Scholar]
  • 25.Brouwers AH, Mulders PF, de Mulder PH, van den Broek WJ, Buijs WC, Mala C, Joosten FB, Oosterwijk E, Boerman OC, Corstens FH, Oyen WJ. Lack of efficacy of two consecutive treatments of radioimmunotherapy with 131I-cG250 in patients with metastasized clear cell renal cell carcinoma. J Clin.Oncol. 2005;23:6540–6548. doi: 10.1200/JCO.2005.07.732. [DOI] [PubMed] [Google Scholar]
  • 26.Brouwers AH, Buijs WC, Oosterwijk E, Boerman OC, Mala C, de Mulder PH, Corstens FH, Mulders PF, Oyen WJ. Targeting of metastatic renal cell carcinoma with the chimeric monoclonal antibody G250 labeled with (131)I or (111)In: an intrapatient comparison. Clin.Cancer Res. 2003;9:3953S–3960S. [PubMed] [Google Scholar]
  • 27.Brouwers AH, Mulders PF, Oyen WJ. Carbonic anhydrase IX expression in clear cell renal cell carcinoma and normal tissues: experiences from (radio) immunotherapy. J Clin.Oncol. 2008;26:3808–3809. doi: 10.1200/JCO.2008.17.6073. [DOI] [PubMed] [Google Scholar]
  • 28.Divgi CR, Pandit-Taskar N, Jungbluth AA, Reuter VE, Gonen M, Ruan S, Pierre C, Nagel A, Pryma DA, Humm J, Larson SM, Old LJ, Russo P. Preoperative characterisation of clear-cell renal carcinoma using iodine-124-labelled antibody chimeric G250 (124I-cG250) and PET in patients with renal masses: a phase I trial. Lancet Oncol. 2007;8:304–310. doi: 10.1016/S1470-2045(07)70044-X. [DOI] [PubMed] [Google Scholar]
  • 29.Pastorekova S, Ratcliffe PJ, Pastorek J. Molecular mechanisms of carbonic anhydrase IX-mediated pH regulation under hypoxia. BJU.Int. 2008;101 Suppl 4:8–15. doi: 10.1111/j.1464-410X.2008.07642.x. [DOI] [PubMed] [Google Scholar]
  • 30. Lawrentschuk N, Lee FT, Jones G, Rigopoulos A, Mountain A, O'Keefe G, Papenfuss AT, Bolton DM, Davis ID, Scott AM. Investigation of hypoxia and carbonic anhydrase IX expression in a renal cell carcinoma xenograft model with oxygen tension measurements and (124)I-cG250 PET/CT. Urol Oncol. 2009 doi: 10.1016/j.urolonc.2009.03.028. •• PET studies demonstrating excellent localization of 124 I-cG250 in tumors in a human RCC xenograft model.
  • 31.Divgi CR, Bander NH, Scott AM, O'Donoghue JA, Sgouros G, Welt S, Finn RD, Morrissey F, Capitelli P, Williams JM, Deland D, Nakhre A, Oosterwijk E, Gulec S, Graham MC, Larson SM, Old LJ. Phase I/II radioimmunotherapy trial with iodine-131-labeled monoclonal antibody G250 in metastatic renal cell carcinoma. Clin.Cancer Res. 1998;4:2729–2739. [PubMed] [Google Scholar]
  • 32.Steffens MG, Boerman OC, Oosterwijk-Wakka JC, Oosterhof GO, Witjes JA, Koenders EB, Oyen WJ, Buijs WC, Debruyne FM, Corstens FH, Oosterwijk E. Targeting of renal cell carcinoma with iodine-131-labeled chimeric monoclonal antibody G250. J Clin.Oncol. 1997;15:1529–1537. doi: 10.1200/JCO.1997.15.4.1529. [DOI] [PubMed] [Google Scholar]
  • 33.Divgi CR, O'Donoghue JA, Welt S, O'Neel J, Finn R, Motzer RJ, Jungbluth A, Hoffman E, Ritter G, Larson SM, Old LJ. Phase I clinical trial with fractionated radioimmunotherapy using 131I-labeled chimeric G250 in metastatic renal cancer. J Nucl Med. 2004;45:1412–1421. [PubMed] [Google Scholar]
  • 34.Brouwers AH, van Eerd JE, Frielink C, Oosterwijk E, Oyen WJ, Corstens FH, Boerman OC. Optimization of radioimmunotherapy of renal cell carcinoma: labeling of monoclonal antibody cG250 with 131I, 90Y, 177Lu, or 186Re. J Nucl Med. 2004;45:327–337. [PubMed] [Google Scholar]
  • 35.Stillebroer AB, Oosterwijk E, Mulders PF, Oyen WJ, Boerman O. Radioimmunotherapy with luthetium-177 labeled monoclonal antibody cG250 in patients with advanced renal cell carcinoma. [abstract] Cancer Biother.Radiopharm. 2008;23:523–524. [Google Scholar]
  • 36. Lassi K, Dawson NA. Emerging therapies in castrate-resistant prostate cancer. Curr OpinOncol. 2009;21:260–265. doi: 10.1097/CCO.0b013e32832a1868. • The review discusses recent developments in the treatment of metastatic hormone-refractory prostate cancer
  • 37. Turkbey B, Albert PS, Kurdziel K, Choyke PL. Imaging localized prostate cancer: current approaches and new developments. AJR Am.J Roentgenol. 2009;192:1471–1480. doi: 10.2214/AJR.09.2527. •• The authors review conventional imaging methods as well as targeted imaging approaches with novel tracers used in the clinical management of prostate cancer
  • 38.Ross JS, Gray KE, Webb IJ, Gray GS, Rolfe M, Schenkein DP, Nanus DM, Millowsky MI, Bander NH. Antibody-based therapeutics: focus on prostate cancer. Cancer Metastasis Rev. 2005;24:521–537. doi: 10.1007/s10555-005-6194-0. [DOI] [PubMed] [Google Scholar]
  • 39.Emonds KM, Swinnen JV, Mortelmans L, Mottaghy FM. Molecular imaging of prostate cancer. Methods. 2009;48:193–199. doi: 10.1016/j.ymeth.2009.03.021. [DOI] [PubMed] [Google Scholar]
  • 40. Bouchelouche K, Capala J, Oehr P. Positron emission tomography/computed tomography and radioimmuotherapy of prostate cancer. Curr Opin Oncol. 2009;21:469–474. doi: 10.1097/CCO.0b013e32832d56e4. • The review focuses on recent advancements in PET/CT and radioimmunotherapy of prostate cancer
  • 41. Elsasser-Beile U, Buhler P, Wolf P. Targeted therapies for prostate cancer against the prostate specific membrane antigen. Curr Drug Targets. 2009;10:118–125. doi: 10.2174/138945009787354601. • The review provides an overview of the new strategies with targeted therapies for prostate cancer against the prostate specific membrane antigen.
  • 42.Smith-Jones PM. Radioimmunotherapy of prostate cancer. Q.J Nucl Med Mol.Imaging. 2004;48:297–304. [PubMed] [Google Scholar]
  • 43.Smith-Jones PM, Vallabahajosula S, Goldsmith SJ, Navarro V, Hunter CJ, Bastidas D, Bander NH. In vitro characterization of radiolabeled monoclonal antibodies specific for the extracellular domain of prostate-specific membrane antigen. Cancer Res. 2000;60:5237–5243. [PubMed] [Google Scholar]
  • 44.McDevitt MR, Barendswaard E, Ma D, Lai L, Curcio MJ, Sgouros G, Ballangrud AM, Yang WH, Finn RD, Pellegrini V, Geerlings MW, Jr, Lee M, Brechbiel MW, Bander NH, Cordon-Cardo C, Scheinberg DA. An alpha-particle emitting antibody ([213Bi]J591) for radioimmunotherapy of prostate cancer. Cancer Res. 2000;60:6095–6100. [PubMed] [Google Scholar]
  • 45.Smith-Jones PM, Vallabhajosula S, Navarro V, Bastidas D, Goldsmith SJ, Bander NH. Radiolabeled monoclonal antibodies specific to the extracellular domain of prostate-specific membrane antigen: preclinical studies in nude mice bearing LNCaP human prostate tumor. J Nucl Med. 2003;44:610–617. [PubMed] [Google Scholar]
  • 46.Olson WC, Heston WD, Rajasekaran AK. Clinical trials of cancer therapies targeting prostate-specific membrane antigen. Rev.Recent Clin.Trials. 2007;2:182–190. doi: 10.2174/157488707781662724. [DOI] [PubMed] [Google Scholar]
  • 47.Tagawa S, Jeske S, Milowski MI, Vallabahajosula S, Goldsmith SJ, Matulich D, Kaplan I, Bander NH, Nanus DM. Phase II trial of 177Lutetium radiolabeled anti-prostate-specific membrane antigen (PSMA) monoclonal antibody J591 (177Lu-J591) in patients with metastatic castrate-resistant prostate cancer [abstract] Cancer Biother.Radiopharm. 2008;23:525. [Google Scholar]
  • 48.Pandit-Taskar N, O'Donoghue JA, Morris MJ, Wills EA, Schwartz LH, Gonen M, Scher HI, Larson SM, Divgi CR. Antibody mass escalation study in patients with castration-resistant prostate cancer using 111In-J591: lesion detectability and dosimetric projections for 90Y radioimmunotherapy. J Nucl Med. 2008;49:1066–1074. doi: 10.2967/jnumed.107.049502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Elsasser-Beile U, Wolf P, Gierschner D, Buhler P, Schultze-Seemann W, Wetterauer U. A new generation of monoclonal and recombinant antibodies against cell-adherent prostate specific membrane antigen for diagnostic and therapeutic targeting of prostate cancer. Prostate. 2006;66:1359–1370. doi: 10.1002/pros.20367. [DOI] [PubMed] [Google Scholar]
  • 50. Wolf P, Freudenberg N, Buhler P, Alt K, Schultze-Seemann W, Wetterauer U, Elsasser-Beile U. Three conformational antibodies specific for different PSMA epitopes are promising diagnostic and therapeutic tools for prostate cancer. Prostate. 2009 doi: 10.1002/pros.21090. • Three new antibodies binding to different extracellular PSMA epitopes.
  • 51. Elsasser-Beile U, Reischl G, Wiehr S, Buhler P, Wolf P, Alt K, Shively J, Judenhofer MS, Machulla HJ, Pichler BJ. PET imaging of prostate cancer xenografts with a highly specific antibody against the prostate-specific membrane antigen. J Nucl Med. 2009;50:606–611. doi: 10.2967/jnumed.108.058487. •• This in vivo PET study demonstrates high and specific uptake of 64Cu-labeled mAb 3/A12 in PSMA positive tumors.
  • 52.Wu AM, Senter PD. Arming antibodies: prospects and challenges for immunoconjugates. Nat.Biotechnol. 2005;23:1137–1146. doi: 10.1038/nbt1141. [DOI] [PubMed] [Google Scholar]
  • 53.Mease RC, Dusich CL, Foss CA, Ravert HT, Dannals RF, Seidel J, Prideaux A, Fox JJ, Sgouros G, Kozikowski AP, Pomper MG. N-[N-[(S)-1,3-Dicarboxypropyl]carbamoyl]-4-[18F]fluorobenzyl-L-cysteine, [18F]DCFBC: a new imaging probe for prostate cancer. Clin.Cancer Res. 2008;14:3036–3043. doi: 10.1158/1078-0432.CCR-07-1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Foss CA, Mease RC, Fan H, Wang Y, Ravert HT, Dannals RF, Olszewski RT, Heston WD, Kozikowski AP, Pomper MG. Radiolabeled small-molecule ligands for prostate-specific membrane antigen: in vivo imaging in experimental models of prostate cancer. Clin.Cancer Res. 2005;11:4022–4028. doi: 10.1158/1078-0432.CCR-04-2690. [DOI] [PubMed] [Google Scholar]
  • 55.Chen Y, Foss CA, Byun Y, Nimmagadda S, Pullambhatla M, Fox JJ, Castanares M, Lupold SE, Babich JW, Mease RC, Pomper MG. Radiohalogenated prostate-specific membrane antigen (PSMA)-based ureas as imaging agents for prostate cancer. J Med Chem. 2008;51(24):7933–7943. doi: 10.1021/jm801055h. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Banerjee SR, Foss CA, Castanares M, Mease RC, Byun Y, Fox JJ, Hilton J, Lupold SE, Kozikowski AP, Pomper MG. Synthesis and evaluation of technetium-99m- and rhenium-labeled inhibitors of the prostate-specific membrane antigen (PSMA) J Med Chem. 2008;51:4504–4517. doi: 10.1021/jm800111u. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Hillier SM, Maresca KP, Femia FJ, Marquis JC, Foss CA, Nguyen N, Zimmerman CN, Barrett JA, Eckelman WC, Pomper MG, Joyal JL, Babich JW. Preclinical evaluation of novel glutamate-urea-lysine analogues that target prostate-specific membrane antigen as molecular imaging pharmaceuticals for prostate cancer. Cancer Res. 2009;69:6932–6940. doi: 10.1158/0008-5472.CAN-09-1682. •• Two novel imaging compounds demonstrate promising results for SPECT/CT imaging of PSMA positive tumors in an in vivo study
  • 58.Jakobovits A. Monoclonal antibody therapy for prostate cancer. Handb.Exp.Pharmacol. 2008;181:237–256. doi: 10.1007/978-3-540-73259-4_11. [DOI] [PubMed] [Google Scholar]
  • 59.Vallabhajosula S, Goldsmith SJ, Hamacher KA, Kostakoglu L, Konishi S, Milowski MI, Nanus DM, Bander NH. Prediction of myelotoxicity based on bone marrow radiation-absorbed dose: radioimmunotherapy studies using 90Y- and 177Lu-labeled J591 antibodies specific for prostate-specific membrane antigen. J Nucl Med. 2005;46:850–858. [PubMed] [Google Scholar]
  • 60.Bander NH, Milowsky MI, Nanus DM, Kostakoglu L, Vallabhajosula S, Goldsmith SJ. Phase I trial of 177lutetium-labeled J591, a monoclonal antibody to prostate-specific membrane antigen, in patients with androgen-independent prostate cancer. J Clin.Oncol. 2005;23:4591–4601. doi: 10.1200/JCO.2005.05.160. [DOI] [PubMed] [Google Scholar]
  • 61.Tolmachev V. Imaging of HER-2 overexpression in tumors for guiding therapy. Curr Pharm.Des. 2008;14:2999–3019. doi: 10.2174/138161208786404290. [DOI] [PubMed] [Google Scholar]
  • 62.Shariat SF, Bensalah K, Karam JA, Roehrborn CG, Gallina A, Lotan Y, Slawin KM, Karakiewicz PI. Preoperative plasma HER2 and epidermal growth factor receptor for staging and prognostication in patients with clinically localized prostate cancer. Clin.Cancer Res. 2007;13:5377–5384. doi: 10.1158/1078-0432.CCR-07-0330. [DOI] [PubMed] [Google Scholar]
  • 63.Shi Y, Brands FH, Chatterjee S, Feng AC, Groshen S, Schewe J, Lieskovsky G, Cote RJ. Her-2/neu expression in prostate cancer: high level of expression associated with exposure to hormone therapy and androgen independent disease. J Urol. 2001;166:1514–1519. doi: 10.1016/s0022-5347(05)65822-3. [DOI] [PubMed] [Google Scholar]
  • 64.Domingo-Domenech J, Fernandez PL, Filella X, Martinez-Fernandez A, Molina R, Fernandez E, Alcaraz A, Codony J, Gascon P, Mellado B. Serum HER2 extracellular domain predicts an aggressive clinical outcome and biological PSA response in hormone-independent prostate cancer patients treated with docetaxel. Ann.Oncol. 2008;19:269–275. doi: 10.1093/annonc/mdm490. [DOI] [PubMed] [Google Scholar]
  • 65.Hansel DE, Swain E, Dreicer R, Tubbs RR. HER2 overexpression and amplification in urothelial carcinoma of the bladder is associated with MYC coamplification in a subset of cases. Am.J Clin.Pathol. 2008;130:274–281. doi: 10.1309/41VLTFX3YPP1HF6F. [DOI] [PubMed] [Google Scholar]
  • 66.Lae M, Couturier J, Oudard S, Radvanyi F, Beuzeboc P, Vieillefond A. Assessing HER2 gene amplification as a potential target for therapy in invasive urothelial bladder cancer with a standardized methodology: results in 1005 patients. Ann.Oncol. 2009 Nov 4; doi: 10.1093/annonc/mdp488. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Caner V, Turk NS, Duzcan F, Tufan NL, Kelten EC, Zencir S, Dodurga Y, Bagci H, Duzcan SE. No strong association between HER-2/neu protein overexpression and gene amplification in high-grade invasive urothelial carcinomas. Pathol.Oncol Res. 2008;14:261–266. doi: 10.1007/s12253-008-9027-y. [DOI] [PubMed] [Google Scholar]
  • 68. McLarty K, Cornelissen B, Cai Z, Scollard DA, Costantini DL, Done SJ, Reilly RM. Micro-SPECT/CT with 111In-DTPA-pertuzumab sensitively detects trastuzumab-mediated HER2 downregulation and tumor response in athymic mice bearing MDA-MB-361 human breast cancer xenografts. J Nucl Med. 2009;50:1340–1348. doi: 10.2967/jnumed.109.062224. •• In this study 111In-DTPA-pertuzumab imaged HER2 downregulation after treatment with trastuzumab in human breast cancer xenografts.
  • 69.Ahlgren S, Wallberg H, Tran TA, Widstrom C, Hjertman M, Abrahmsen L, Berndorff D, Dinkelborg LM, Cyr JE, Feldwisch J, Orlova A, Tolmachev V. Targeting of HER2-expressing tumors with a site-specifically 99mTc-labeled recombinant affibody molecule, ZHER2:2395, with C-terminally engineered cysteine. J Nucl Med. 2009;50:781–789. doi: 10.2967/jnumed.108.056929. [DOI] [PubMed] [Google Scholar]
  • 70.Smith-Jones PM, Solit D, Afroze F, Rosen N, Larson SM. Early tumor response to Hsp90 therapy using HER2 PET: comparison with 18F-FDG PET. J Nucl Med. 2006;47:793–796. [PMC free article] [PubMed] [Google Scholar]
  • 71.Cheng Z, De Jesus OP, Namavari M, De A, Levi J, Webster JM, Zhang R, Lee B, Syud FA, Gambhir SS. Small-animal PET imaging of human epidermal growth factor receptor type 2 expression with site-specific 18F-labeled protein scaffold molecules. J Nucl Med. 2008;49:804–813. doi: 10.2967/jnumed.107.047381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Orlova A, Wallberg H, Stone-Elander S, Tolmachev V. On the selection of a tracer for PET imaging of HER2-expressing tumors: direct comparison of a 124I-labeled Affibody molecule and trastuzumab in a murine xenograft model. J Nucl Med. 2009;50:417–425. doi: 10.2967/jnumed.108.057919. • Affibody molecules may be superior to trastuzumab for PET imaging of HER2 positive tumors.
  • 73.Kramer-Marek G, Kiesewetter DO, Martiniova L, Jagoda E, Lee SB, Capala J. [18F]FBEM-Z(HER2:342)-Affibody molecule-a new molecular tracer for in vivo monitoring of HER2 expression by positron emission tomography. Eur.J Nucl Med Mol.Imaging. 2008;35:1008–1018. doi: 10.1007/s00259-007-0658-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Kramer-Marek G, Kiesewetter DO, Capala J. Changes in HER2 expression in breast cancer xenografts after therapy can be quantified using PET and 18F-labeled Affibody molecules. J Nucl Med. 2009;50:1131–1139. doi: 10.2967/jnumed.108.057695. •• The study demonstrates that 18F-labelled Affibody molecules are useful for PET imaging of HER2 that allows monitoring of expression levels and quantification of possible changes in receptor expression due to therapeutic intervention and, thereby, may provide means for early assessment of tumour response.
  • 75.Baum R, Orlova A, Tolmachev V, Feldwisch J. A novel molecular imaging agent for diagnosis of recurrent HER2 positive breast cancer. First time in human study using Indium-111- or Galium-68-labelled Affibody molecule. [abstract] Eur J Nucl Med Mol.Imaging. 2006;33:S91. [Google Scholar]
  • 76.Tolmachev V, Orlova A, Pehrson R, Galli J, Baastrup B, Andersson K, Sandstrom M, Rosik D, Carlsson J, Lundqvist H, Wennborg A, Nilsson FY. Radionuclide therapy of HER2-positive microxenografts using a 177Lu-labeled HER2-specific Affibody molecule. Cancer Res. 2007;67:2773–2782. doi: 10.1158/0008-5472.CAN-06-1630. [DOI] [PubMed] [Google Scholar]
  • 77. Orlova A, Tran TA, Ekblad T, Karlstrom AE, Tolmachev V. (186)Re-maSGS-Z (HER2:342), a potential Affibody conjugate for systemic therapy of HER2-expressing tumours. Eur J Nucl Med Mol.Imaging. 2009 Sep 22; doi: 10.1007/s00259-009-1268-9. [Epub ahead of print] • Rhenium-labeled Affibody molecules are potential radiotherapeutic agents for radionuclide therapy of HER2 positive tumors.
  • 78.Ballangrud AM, Yang WH, Palm S, Enmon R, Borchardt PE, Pellegrini VA, McDevitt MR, Scheinberg DA, Sgouros G. Alpha-particle emitting atomic generator (Actinium-225)-labeled trastuzumab (herceptin) targeting of breast cancer spheroids: efficacy versus HER2/neu expression. Clin.Cancer Res. 2004;10:4489–4497. doi: 10.1158/1078-0432.CCR-03-0800. [DOI] [PubMed] [Google Scholar]
  • 79.Costantini DL, Bateman K, McLarty K, Vallis KA, Reilly RM. Trastuzumab-resistant breast cancer cells remain sensitive to the auger electron-emitting radiotherapeutic agent 111In-NLS-trastuzumab and are radiosensitized by methotrexate. J Nucl Med. 2008;49:1498–1505. doi: 10.2967/jnumed.108.051771. [DOI] [PubMed] [Google Scholar]

RESOURCES