Abstract
Only a handful of radiolabeled antibodies (Abs) have gained FDA approval for use in clinical oncology, including four immunodiagnostic agents and two targeted radioimmunotherapeutic agents. Despite the advent of non-immunogenic Abs and availability of a diverse library of radionuclides, progress beyond early Phase II RIT studies in solid tumors has been marginal. Furthermore, 18F-FDG continues to dominate the molecular imaging domain, underscored by a decade-long absence of any newly approved antibody-based imaging agents (none since 1996!). Why has the development of clinically successful Abs for RIT been limited to lymphoma? What obstacles must be overcome to allow the FDA-approval of immunoPET imaging agents? How can we address the unique challenges that have thus far prevented the introduction of Ab-based imaging agents and therapeutics for solid tumors? Many poor decisions have been made regarding radiolabeled Abs, but useful insight can be gained from these mistakes. The following review addresses physical, chemical, biological, clinical, regulatory, and financial limitations that impede the progress of this increasingly important class of drugs.
Overview
The following review illustrates key components of a successful radiolabeled diagnostic or therapeutic antibody (Ab) from the inside out — that is, starting from the unstable nucleus itself and moving outwards. For each sequential topic, relevant theory will be examined and related to the practical use of various radiolabeled Abs that have succeeded to varying degrees in the clinic. This approach will present each important aspect of a rather complex multidisplinary phenomenon in a logical, stepwise manner:
-
The radionuclide itself
Physical properties of the unstable nucleus
-
The radionuclide's chemical surroundings
Chemical attachment of the radiometal or radiohalogen
-
The antibody
Biological issues of the radioimmunoconjugate
-
The target antigen and associated tumor(s)
Clinical efficacy of a given target receptor or antigen
-
Challenges and Prospects
Overcoming failures through wiser choices
The inside-out theme is also preserved within Section I–Section IV by initially analyzing the scientific efforts inside the laboratory, followed by a glimpse out into to the real world, where commercial, regulatory, and financial obstacles are encountered that impede the accessibility of drugs to the target patient population.
I. The radionuclide itself
Radionuclides in Targeted Radioimmunotherapy
The commercially successful Genentech/Roche antibodies (Abs) (e.g. rituximab, trastuzumab, and bevasizumab) and most of the monoclonal Abs on the market or in late-stage development are naked, or unconjugated Abs that function by targeting tumor-expressed proteins. An alternative strategy is to use the Abs for targeted delivery of a cytotoxic drug or radionuclide thereby enhancing the therapeutic efficacy of the Ab.1–4
Ehrlich conceived the idea of "magic bullets" targeting compounds and eradicating disease5, but it was not until the early 1950s that an Ab was conjugated to a radionuclide.6 Pioneering clinical studies by Mach7 and Goldenberg8 with anti-CEA Abs demonstrated the feasibility of specific targeting. Kohler isolated the first monoclonal antibodies in 1975.9 Nearly two decades later, Leichner and co-workers studied the use of 111In/90Y-labeled anti-ferritin for RIT in patients with hepatoma.10 Finally, the FDA approvals for two radiolabeled anti-CD20 mAbs, 90Y-labeled Zevalin® (ibritumomab tiuxetan) in 2002 and 131I-labeled Bexxar® in 2003 for the treatment of non-Hodgkin’s lymphoma (NHL) were landmark events in the developmental history of therapeutic radiolabeled mAbs (RIT) (see Section IV).11
Radionuclides in Molecular Imaging
Molecular imaging is a rapidly emerging field and a powerful tool in the clinical diagnosis of disease.12 Radioimmunoimaging has been traditionally developed in parallel with RIT for evaluating targeting and dosimetry. MAb-based tracers are gaining acceptance for identification of specific molecular targets and visualization of tumors at primary and metastatic sites. Drug development is also being revolutionized by molecular imaging probes, especially in the field of oncology.13
The most sensitive imaging modalities utilize radiotracers, including gamma (γ)- camera (planar) scintigraphy, single-photon emission computerized tomography (SPECT), and positron emission tomography (PET) with PET operating at 10-fold sensitivity superiority to SPECT. Radioactive modalities coupled with non-radioactive modalities such as optical imaging, magnetic resonance imaging (MRI), and computed tomography (CT) provide multimodal imaging approches with significant advantages combining the strengths of complementary modalities (e.g., PET-CT couples high sensitivity of PET with detailed anatomical information from CT).
Inherent Nuclear Properties: Half-Life, Decay Energy, and Range
A successful diagnostic or radio immunotherapeutic Ab must be radiolabeled with a radionuclide matched for the intended use.14, 15 The half-life (t½) must be adequate for target accumulation and non-specific clearance. Considering these requirements, labeling an intact IgG with 18F is an impractical endeavor. Copper-64 (t½ ~ 12 h) would be a more reasonable choice for radiolabeling antibodies .16–18 On this basis, 89Zr (t½ ~ 3.3 d) is perhaps even better suited for immunoPET.19–23 Several other radionuclides (Table 1) also possess half-lives compatible with mAb biological half-lives.
Table 1.
Physical Data of useful Radionuclides for Immunodiagnosis (γ/β+) and for RIT (β−/β+/α/Auger). Information was obtained from Kocher175 (1981) and Milenic et al1 (2004).
Radio-nuclide | t½ (h) | Imaging Decay | RIT Decay | Rmean† (mm) | Eα (keV) | Eβ [avg] (keV) | Eγ (keV) | Production |
---|---|---|---|---|---|---|---|---|
90Y | 64.08 | ---- | β− | 3.78 | ---- | 934.8 | ---- | 90Sr/90Y generator |
89Y(n,γ)90Y | ||||||||
131I | 192.96 | γ | β− | 0.36 | ---- | 181.7 | 364.5 | 131Te(n,γ)131I |
177Lu | 161.04 | γ | β− | 0.22 | ---- | 133.0 | 208.4 | 176Lu(n,γ)177Lu |
EC | <0.02 | 113.0 | ||||||
67Cu | 61.92 | γ | β− | 0.24 | ---- | 141.0 | 184.6 | 68Zn(p,2p)67Cu |
EC | <0.02 | 93.3 | 67Zn(n,p)67Cu | |||||
91.3 | ||||||||
166Ho | 26.80 | γ | β− | 2.43 | ---- | 665.7 | 80.6 | 166Dy/166Ho generator |
EC | <0.02 | 1379.4 | ||||||
186Re | 90.64 | γ | β− | 0.98 | ---- | 349.3 | 137.1 | 185Re(n,γ)186Re |
EC | <0.02 | 122.3 | ||||||
188Re | 16.98 | γ | β− | 2.91 | ---- | 764.2 | 155 | 188W/188Re generator |
EC | <0.02 | 633.1 | 187Re(n,γ)188Re | |||||
153Sm | 46.27 | γ | β− | 0.50 | ---- | 223.6 | 103.2 | 152Sm(n,γ)153Sm |
111Ag | 179.28 | γ | β− | 0.98 | ---- | 349.8 | 342.1 | 110Pd(n,γ)111Pd(β−)→ |
111mAg(γ)111Ag | ||||||||
109Pd | 13.45 | ---- | β− | 1.03 | ---- | 361.0 | ---- | 108Pd(n,γ)109Pd |
165Dy | 2.33 | γ | β− | 1.37 | ---- | 440.2 | 94.7 | 164Dy(n,γ)165Dy |
EC | <0.02 | |||||||
166Dy | 81.6 | γ | β− | 0.18 | ---- | 118.3 | 82.47 | 164Dy(n,γ)165Dy |
EC | <0.02 | 165Dy(n,γ)166Dy | ||||||
169Er | 225.6 | γ | β− | 0.13 | ---- | 99.4 | 8.4 | 168Er(n,γ)169Er |
169Tm(n,p)169Er | ||||||||
175Yb | 100.56 | γ | β− | 0.20 | ---- | 125.5 | 369.3 | 174Yb(n,γ)175Yb |
EC | <0.02 | 282.5 | ||||||
198Au | 64.70 | γ | β− | 0.83 | ---- | 311.5 | 411.8 | 197Au(n,γ)198Au |
EC | <0.02 | 675.9 | ||||||
212Pb | 10.64 | γ | β− | 0.13 | ---- | 99.4 | 238.6 | 224Ra/212Pb generator |
EC | <0.02 | |||||||
212Bi | 1.01 | γ | α | 0.04–0.1 | 6,090 | 717.3 | 727.2 | 212Pb/212Bi generator |
β− | 2.68 | 6,051 | 39.8 | 228Th/212Bi generator | ||||
EC | <0.02 | |||||||
213Bi | 0.76 | γ | α | 0.04–0.1 | 5,870 | 430.0 | 440.4 | 225Ac/213Bi generator |
β− | 1.32 | 5,549 | ||||||
EC | <0.02 | |||||||
225Ac | 240.00 | γ | α | 0.04–0.1 | 5,829 | ---- | 99.8 | n-capture of 232Th → |
EC | <0.02 | 5,792 | 86.1 | 233U → 225Ac or | ||||
5,731 | 226Ra(p,2n)225Ac | |||||||
211At | 7.21 | γ | α | 0.04–0.1 | 5,867 | ---- | 687.0 | 209Bi(α,2n)211At |
EC | <0.02 | |||||||
149Tb | 4.12 | β+, γ | α | 0.04–0.1 | 3,967 | 921.0 | 352.2 | (Ta(p,spall) |
β+ | 3.71 | 165.0 | 152Gd(p,4n)149Tb or | |||||
EC | <0.02 | 3883. | 142Nd(12C,5n)149Tb | |||||
195mPt | 96.48 | γ | IT/EC | <0.02 | ---- | ---- | 98.9 | 195Pt(n,n’)195mPt |
129.8 | 194Pt(n,γ)195mPt | |||||||
203Pb | 52.02 | γ | EC | <0.02 | ---- | ---- | 279.2 | 203Tl(p,n)203Pb |
401.3 | 203Tl(d,2n)203Pb | |||||||
201Tl | 73.06 | γ | EC | <0.02 | ---- | ---- | 167.4 | 203Tl(p,3n)201Pb201Tl |
135.3 | ||||||||
125I | 1443.4 | γ | EC | <0.02 | ---- | ---- | 35.49 | 124Xe(n,γ)125Xe/125I |
67Ga | 78.26 | γ | EC | <0.02 | ---- | ---- | 93.3 | 68Zn(p,2n)67Ga |
184.6 | ||||||||
300.2 | ||||||||
111In | 67.31 | γ | EC | <0.02 | ---- | ---- | 171.3 | 111Cd(p,n)111In |
245.4 | ||||||||
153Gd | 5,798 | γ | EC | <0.02 | ---- | ---- | 97.4 | Eu(n,γ) 152Eu/152Gd |
103.2 | 152Gd (n,γ)153Gd | |||||||
123I | 13.13 | γ | EC | <0.02 | ---- | ---- | 159.0 | 124Te(p,2n)123I |
124I | 100.22 | β+, γ | β+ | 3.25 | ---- | 830.5 | 602.7 | 214Te(p,n)124I |
EC | <0.02 | |||||||
66Ga | 9.40 | β+,γ | β+ | 8.06 | ---- | 1739.1 | 1039.3 | 66Zn(p,n)66Ga |
2752.1 | ||||||||
55Co | 17.53 | β+, γ | β+ | 5.74 | ---- | 1306.8 | 1408.4 | 54Fe(d,n)55Co |
EC | <0.02 | 477.2 | 56Fe(p,2n)55Co | |||||
931.2 | ||||||||
77Br | 57.04 | β+,γ | β+ | 0.27 | ---- | 151.7 | 238.9 | 77Se(p,n)77Br |
EC | <0.02 | 520.7 | ||||||
76Br | 16.00 | β+,γ | β+ | 5.07 | ---- | 1180 | 559.1 | 76Se(p,n)76Br |
EC | <0.02 | 657.0 | ||||||
1853.7 | ||||||||
89Zr | 78.43 | β+,γ | β+ | 1.18 | ---- | 396.9 | 909.1 | 89Y(p,n)89Zr |
EC | <0.02 | |||||||
64Cu | 12.70 | β+,γ | β+ | 0.70 | ---- | β+ 278.1 | 1345.8 | 64Ni(p,2p)64Cu |
β− | 0.39 | β− 190.2 | ||||||
EC | <0.02 | |||||||
86Y | 14.74 | β+,γ | β+ | 2.46 | ---- | 672 | 1076.7 | 86Sr(p,n)86Y |
EC | <0.02 | 627.8 | ||||||
1153.1 | ||||||||
99mTc | 6.02 | γ | IT/EC | ---- | ---- | ---- | 140.5 | 99Mo/99mTc generator |
Mean β ranges (Rmean, mm) were calculated from mean β energies (Eβ [avg], MeV) using the following equation 176: Rmean = 4.12 · Emean1.265 − 0.094 · lnEmean
Radionuclide emissions are critical in the design of a given imaging or therapeutic application. Early RIT studies used 131I due to advantages of commercial availability, potential direct dosimetry, and simple familiarity. However, the t½ is unnecessarily long, the γ-emissions require patient isolation, and absorbed doses are compromised by a short biological half-life resulting from dehalogenation within blood and tumor sites.
Methods for attaching metal chelating groups have facilitated study of metallic radionuclides (e.g., 90Y, 177Lu, and 67Cu) that are potentially far better suited for RIT. Yttrium-90 lacks interfering γ-emissions altogether, and, while 177Lu and 67Cu emit γ-rays, they are of much lower energy with respect to 131I. Yttrium-90 provides advantages over 131I because it delivers on average a more energetic tumor-killing β− (935 keV versus 182 keV for 131I) and concomitantly, a longer mean range (3.78 mm versus 0.36 mm for 131I) (Table 1). These characteristics improve the ability of a radiolabeled Ab to kill both targeted and neighboring cells and offer a particular advantage in treating bulky or poorly vascularized disease. In addition, being a pure β-emitting radionuclide, 90Y can be administered on an outpatient basis.
Radionuclide characteristics affect dose deposition. Lutetium-177 is not optimal for treatment of a large-diameter solid tumor as low energy β particles lack sufficient tumor penetration while conversely, 90Y is not suited for treating disseminated microscopic tumor burden. Use of 131I, 177Lu, or 67Cu is more appropriate in an adjuvant or small tumor setting. In contrast, 90Y seems suited for use in patients with clearly detectable lesions due to its higher energy and consequent better penetration. Successful matching of β-emission energies to tumor volumes in RIT has been well defined.24, 25
Choosing an appropriate radionuclide is equally important for imaging applications. For example, a high energy positron (β+) emission (e.g. 66Ga) may produce PET images of relatively poor quality and resolution; lower energy β+ emitters (e.g. 64Cu) are more desirable.16 Indium-111 is a popular choice for SPECT imaging. Lower β+ and γ energies are associated with higher PET and SPECT image quality, respectively.
Examples of matched β+/β− (imaging/therapy) pairs include 124I/131I, 86Y/90Y, and 64Cu/67Cu. Bremsstrahlung imaging of 90Y is sub-optimal, and although 111In can be used as a surrogate, PET imaging using 86Y provides the advantage of matched chemistry. Associated β−, β+ and Auger electron emissions render 64Cu useful for both RIT and PET imaging. More efficacious therapeutic effects may be possible using the β−-emitter 67Cu.26, 27
Radiobiology and Dosimetry
Radionuclide choice is ideally influenced by clinical parameters such as tumor location, size, morphology, physiology, and radiosensitivity. Important variables affecting tumor response include cumulative radiation dose delivered to the tumor(s), dose rate, penetration, and radiosensitivity. The extent of heterogeneity of dose deposition is highly dependent on Ab characteristics and radionuclide properties. Furthermore, enhanced therapeutic efficacy can be attained through selective dose delivery to radiosensitive areas of tumor. The percent of the injected dose per gram of tumor (%ID/g) expresses the tumor uptake of a given agent. Quite encouraging %ID/g values are often observed in rodents, but this trend is deceptively optimistic and does not apply to humans. Tumor uptake and penetration have been the most challenging limitations, with accretion at such low levels as 0.001–0.01 %ID/g often resulting in tumor doses of <1,500 cGy. Bone marrow is typically the dose-limiting organ, with an apparent dose limitation of about 150–200 cGy.
Any particulate emission – including helium nuclei (α particles), negative electrons (negatrons, β− particles), positive electrons, (positrons, β+ particles), and Auger electrons – may be employed in RIT; however, β−-emitters are most often employed for a variety of reasons. A β− particle travels long distances before dissipating all of its kinetic energy, and this low-linear energy transfer (low-LET) emission can span up to 50 cell diameters. Single-strand DNA breaks typically occur in cells traversed by β− particles, and bystander effects on antigen-negative neighboring cells can also be exerted. A unique β− emission energy spectrum — not a discretely defined value — governs the observed path lengths for a given radionuclide. Normal tissue damage is one of the disadvantages associated with β− emissions. Circulation through bone marrow provides opportunity for circulating radionuclide conjugates to irradiate the marrow cells leading to myelosuppression.
An alpha (α) particle has a very short path length (< 100 µm), but a very high LET28, with a typical energy deposition of ~ 100 keV/µm compared to 0.2 keV/µm from a β− emission. The relative biologic effectiveness (RBE) of high-LET radiation exhibits no dose rate dependence and is effective even under hypoxic conditions. An individual cancer cell can be killed by interaction with only a few and possibly with only a single α-particle. Moreover, the range of α particles is short enough to minimize normal tissue damage. Alpha particles (i.e. helium nuclei) are quite large, have high energies of several MeV, and are associated with high probability of inflicting irreparable and cytocidal DNA double-strand breaks.29 Consequently, α-emitters are well suited for hematologic disease, micrometastatic disease, and tumor cells near the surface of cavities.
Alpha-emitting radionuclides with ideal characteristics for RIT include 212Bi 30–34, 225Ac/213Bi 35–42, 211At 43–45, and 149Tb 41, 46 (Table 1). Alpha-targeted therapy using 212Bi or 213Bi may be achieved either directly or by using an in vivo generator through injection of the appropriate parent radionuclide, 212Pb or 225Ac, respectively. An attractive longer-lived α-emitter is 211At, with a t½ of 7.2 hours. The successful application of α-emitters in RIT has awaited developments in radionuclide production, protein labeling chemistry, dosimetry, and the development of appropriate pre-clinical models. A number of preclinical studies have concluded that α-emitters may be more effective than β− emitters administered at comparable doses in RIT.47, 48
Auger and conversion electron emitters’ emissions are highly localized and are only effective post-localization in the cell nucleus.49, 50 Auger emitters have the shortest range of energy deposition, typically submicrometer, that is, less than one cell diameter. Indium-111 releases approximately 7 electrons per decay, whereas 125I and 195mPt release 20–35 electrons per decay. Auger-emitting radionuclides exhibit desireable characteristics, especially under appropriately selected conditions including single-cell systems like leukemia, adjuvant settings, and post-surgical treatment of residual disease.
Commercialization of Radionuclide Production
The commercial availability of radionuclides such as 131I, 125I, 111In, 90Y, and 99mTc has directly contributed to the pace at which diagnostic and therapeutic immunoconjugates have been developed and tested in clinical trials, although the converse may also be true.51 A number of generator-produced radionuclides are readily available (e.g., 188Re, 212Bi, 90Y, 99mTc), making them ideal candidates for widespread hospital-based applications. MDS Nordion has focused on supplying radionuclides for the emerging RIT business. Nordion extended its deal with Corixa to radiolabel the Ab in Bexxar® with 131I, and also produced sterile 90Y for IDEC’s Zevalin®.
Regulatory, Financial, and Commercial Barriers
Radioactive decay, an inherent property that is crucial to the action of both imaging and RIT, is ironically one of the biggest hurdles to overcome. The fear of radioactive materials is a great obstacle that increases commercial production costs and influences public perception, industrial reluctance, and governmental regulation. While the development of non-radioactive cancer Ab immunotherapy is very much a growing industry, seemingly minimal effort is being expended in RIT, except by academic researchers.
Procedural and logistic hurdles in conducting RIT are often substantial.52 MDS Nordion has a unique position at the crossroads of the nuclear and drug research industries, experiencing both sides with stringent and often contradictory regulations. Beyond satisfying FDA guidelines, radiation safety issues abound. In the clinical setting, highly specialized teams are necessary, and for some high-dose protocols, major physical plant radiation shielding alterations can be required. Acquisition and radionuclide use can necessitate hospital-based care, making RIT more complicated than other therapies.
High cost and limited or unresolved availability are major obstacles that have limited the clinical evaluation of Abs radiolabeled with 213Bi, 212Pb, 64Cu, 67Cu, 177Lu, and 86Y. Some β−-emitting radionuclides have not been available in adequate amounts (e.g., 177Lu), while others await the development of improved bifunctional chelators (e.g., 67Cu). Alpha emitters (e.g., 211At, 213Bi) possess great potential for RIT, but face equally complex challenges due to limited production and availability status.
Decisions concerning choice of radionuclide have been made regardless of real appropriateness; selection criteria have instead been based on availability, popularity, familiarity, or historical trends. Limited access to facile radiolabeling chemistry also contributes to this problem. A grand total of only four radionuclides, 111In, 99mTc, 131I, and 90Y, have been integrated into FDA-approved radiolabeled Abs (Table 2). All four of these radionuclides are widely available, relatively inexpensive, and in some small part this status reflects the development and existence of actual products. Two of these radionuclides, 90Y and 131I, are components of approved therapeutics, while 111In and 99mTc are approved for traditional γ scintigraphic imaging. Strikingly, no Abs labeled with β+ emitting (PET) radionuclides have been approved despite the superior image resolution and sensitivity of PET. There are also no approved α-emitting radionuclides which seem obviously far better suited for treating small tumor burden and micrometastases than high-energy β−-emitters. Several promising lower-energy β−-emitters (e.g., 177Lu and 67Cu) also await successful commercialization.
Table 2.
Current FDA-Approved Antibodies for the Parenteral Use in Detection and Treatment of Cancer. Derived from Milenic et al1 (2004) and Walsh93 (2006).
Generic Name | Trade Name | Agent/Target | Cancer Indication | Approval |
---|---|---|---|---|
Unconjugated | ||||
Rituximab | Rituxan® | chimeric anti-CD20 IgG1 | B-cell lymphoma | 1997 |
Trastuzumab | Herceptin® | humanized anti-HER2 IgG1 | breast | 1998 |
Alemtuzumab | CamPath® | humanized anti-CD52 | chromic lymphocytic leukemia | 2001 |
Cetuximab | Erbitux® | chimeric anti-EGFR | colorectal head/neck | 2004 2006 |
Bevacizumab | Avastin® | chimeric anti-VEGF | colorectal | 2004 |
Radioconjugates | ||||
Satumomab pendetide | OncoScint®* | 111In-murine anti-TAG-72 IgG | colorectal, ovarian | 1992 |
Nofetumomab merpentan | Verluma®* | 99mTc-murine anti-EGP-1 Fab' | small cell lung | 1996 |
Arcitumomab | CEA-Scan®* | 99mTc-murine anti-CEA Fab' | colorectal | 1996 |
Capromab pendetide | ProstaScint® | 111In-murine anti-PSMA | prostate | 1996 |
Ibritumomab tiuxetan | Zevalin® | 90Y-murine anti-CD20 IgG + rituximab | B-cell lymphoma | 2002 |
Tositumomab “Anti-B1” | Bexxar® | 131I-murine anti-CD20 IgG + unlabeled tositumomab | B-cell lymphoma | 2003 |
Drug conjugates | ||||
Gemtuzumab ozogamicin | Mylotarg® | humanized anti-CD33 IgG4 conjugated to colicheamicin | acute myelogenous leukemia | 2000 |
No longer commercially available.
Although radionuclide cost can significantly elevate the cost of RIT agents per dose relative to non-radioactive therapies; fewer administered doses of RIT are typically necessary to achieve comparable results. Average costs per year were determined to be $17,529 for 90Y-Zevalin® RIT,53 $23,099 for a four-dose rituximab regimen,54 and $25,559 for an eight-dose rituximab regimen.54 The average durations of remission were 14.4 months with 90Y-Zevalin® RIT,53 6.2 months with a four-dose rituximab regimen,54 and 11.4 months with an eight-dose rituximab regimen.54 In this scenario, clinical data has validated that RIT is both more cost effective and more efficacious than nonradioactive immunotherapy.
Sadly, a serious conflict of interest may prevent cancer patients from receiving appropriate treatment. In some cases, patients are prescribed drugs that are unnecessary or insufficient simply because they are more profitable for the physician. In a nationally televised broadcast, Brian Williams and his colleague, Rehema Ellis, recently addressed this moral and ethical dilemma on the NBC Nightly News on September 21, 2006.55 In the context of RIT, the referring physician (i.e. medical oncologist) may lose profits by referring his patient to a nuclear medicine physician (i.e. radiation oncologist). The referring oncologist, often lacking the proper licensing for injection of radiopharmaceuticals, does not ultimately administer the drug and hence does not gain revenue. The economics of medical fees and billing as structured by modern medicine may determine the choice of administered therapy despite the proven clinical advantages of RIT.
Beyond the argument of cost effectiveness, quality of life issues should be weighed. Non-radioactive immunotherapy or chemotherapy can be followed by relapses, complications, and debilitating side effects, deleteriously impacting emotional health and personal finances. In contrast, RIT treatment is administered in two doses, one week apart, usually with minimal side effects. The real cost of current RIT therapies must be placed in context with financial burden, survival, and quality of life in addition to efficacy.
II. The radionuclide's chemical surroundings
Immediate chemical environment
Ideally, the cytotoxic/imaging agent should be linked to the carrier without impairing functionality of either while maintaining a stable linkage. Stable attachment of radiometals to Abs has been pursued to circumvent the pitfalls of radioiodine — especially dehalogenation of internalizing Abs — that preclude their use for RIT. The benefits of 90Y and 111In over 131I have been demonstrated by clinically comparing tumor uptake kinetics of 90Y- and 131I-labeled Abs and by imaging studies with 111In and 131I.
The clinical application supposedly guides radionuclide choice, which, in turn, determines the radiolabeling procedure. No single radionuclide is suitable for all clinical applications. Likewise, no single chelator is compatible with the full spectrum of available metal ions. Fundamental differences in coordination chemistry define that different metals vary in their preference of ligand.56
Radiolabeling Protocols
Radiolabeling procedures must be simple, efficient, reproducible, and affordable. Currently, only 131I and 90Y appear to fulfill the present requirements for commercial development. Direct radioiodination (131I, 125I, 124I, and 123I) is well established, and tositumomab, the Ab in Bexxar®, is radiolabeled with 131I by this means.
Metallic radionuclides such as 90Y, 111In, 67Cu, and 177Lu require chelation chemistry for radiolabeling to an Ab. Radiometal complexation must occur under practical conditions. Some complexation reactions can be quite slow, requiring elevated temperature and/or high pH values over extended time periods and therefore compromising the use of such radiolabeling methodologies. Generally, DTPA (diethylenetriaminepentaacetic acid) derivatives and other acyclic chelates exhibit fast complex association rates, whereas 1,4,7,20-tetraazacyclododecane N,N’,N”,N’”-tetraacetic acid (DOTA) derivatives and other macrocyclic chelates have slower complex dissociation rates.
Method and degree of connection to the antibody
The conjugation of a radionuclide to an Ab must fulfill a single, critical requirement: maintain the affinity/avidity of the Ab for its target antigen. Chemical linkage methodologies comprised of thiourea, thioether, amide, ester, and disulfide bonds are available. As for the extent of conjugation, an optimal 'happy medium' must be established wherein degree of protein modification is optimized without reducing binding activity. This key variable is generally controlled by adjusting reaction stoichiometry and reaction conditions (e.g., pH, temperature, and time).
Metal Complex Stability
Choosing a bifunctional chelating agent that forms an adequately kinetically and thermodynamically stable complex within the context of radionuclide choice and application is also critical. For example, in Zevalin®, the 90Y is stably chelated by tiuxetan, which is covalently linked to the mAb, ibritumomab, via thiourea bonds to lysine residues of the IgG. Inadequate stability would lead to loss of the radionuclide into inherent proteins in vivo, increasing background in diagnostic imaging and/or imparting radiation doses to normal organs during RIT.
Available bifunctional chelators
Acyclic DTPA-based and macrocyclic DOTA-based chelators represent the most commonly utilized classes of agents used in RIT. While DTPA-analogs boast rapid complexation kinetics for Y(III) and other lanthanides, a compromise is accepted, as in vivo kinetic stabilities are lower than for macrocyclic chelators. This is especially detrimental when using 90Y, which leaches from the DTPA chelate in vivo over time and accumulates in skeletal tissue in close proximity to highly radiosensitive bone marrow, although it is difficult to deconvolute this effect from circulatory exposures. Conversely, DOTA-based chelators tend to exhibit high in vivo stability for lanthanides but require somewhat elevated temperatures and/or longer complexation reaction times.
The structurally reinforced CHX-DTPA family of chelators is a significant improvement over the traditional DTPA chelators. These preorganized chelators form complexes with high in vivo stability without sacrificing rapid complexation kinetics. Furthermore, CHX-A”, a specific stereoisomer of the CHX-DTPA family, is an efficacious carrier of 86Y and 90Y for PET imaging 57 and radiotherapy 58, respectively, and is the premier chelator for stable complexation of 213Bi.59
The development of improved chelators is an active pursuit for non-lanthanides as well. Crossbridged macrocyclic chelators are currently being investigated for Cu(II) complexes, as these complexes are quite inert relative to those formed with either DOTA or 1,4,8,11-tetraacetic acid (TETA), both of which have exhibited in vivo instability with copper radionuclides.60 While the crossbridged cyclam chelator 4,11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecane (CB-TE2A) has shown promising results with peptides, it is ill-suited for Ab radiolabeling due to somewhat harsh radiolabeling requirements.61 Further refinements must be performed to develop an optimal Ab-compatible Cu chelator.
Commercialization of Chelate Production
To date, no company can boast that it has played a primary role in developing a bifunctional chelate for a marketed radiolabeled protein. Bexxar® does not require a chelating agent, and tiuxetan (MXDTPA), the chelating agent used in Zevalin®, was developed years ago originally at the NCI. Established in 2000, Macrocyclics is a small firm that has been developing cGMP capabilities and commercializing chelant technologies emerging from the Sherry lab at the University of Texas, Dallas.
Dow Chemical Company established ChelaMed radiopharmaceutical services to integrate its chelating agent operations into current Good Manufacturing Practices (cGMP)-compliant pharmaceutical manufacturing.62 MeO-DOTA, Dow’s premier chelating agent, can form stable complexes with radiometals like 177Lu or 90Y. Dow has collaborated with Memorial Sloan-Kettering Cancer Center in the development of 225Ac-labeled mAbs targeted to a variety of cancer types. Avidex Limited has applied Dow’s technology to radiolabel Avidex’s monoclonal T cell receptors (mTCRs). Cytogen Corporation has partnered with Dow in the development of Quadramet® (153Sm- EDTMP), a radiopharmaceutical for treatment of pain associated with bone cancer. NeoRX Corporation had an exclusive worldwide license from Dow to 166Ho-DOTMP for bone marrow ablation. Dow has also participated in collaborations with Kereos, Human Genome Sciences (HGS), Proxima Therapeutics, Barnes Jewish Hospital/Washington University School of Medicine, and Michigan Research Institute.
III. The antibody
Nature of origin
Monoclonal Abs were initially derived from mouse B-cell hybridomas. These murine derived Abs have been useful in the in vitro diagnostics market. However, injection into humans as in vivo imaging agents or for therapeutic purposes induces a human anti-mouse antibody (HAMA) response. To minimize HAMA, chimeric Abs were developed through fusion of the variable region of the mouse Ab with the constant region of a human Ab. Further reductions in HAMA were realized by developing humanized Abs retaining only the CDR region of the murine Ab, while more recent methods have generated fully human monoclonal Abs.63
Zevalin® and Bexxar® are of murine origin and are indicated for therapy of lymphoma, a disease characterized by decreased host-immune recognition. In contrast, more severe immunogenicity of murine Abs in solid tumors has precluded repeat administration and hence meaningful therapy. Effective solid tumor RIT can be achieved only through the continual development of non-immunogenic Abs. A compromise is involved, however, as humanized Abs tend to remain in circulation longer, effectively decreasing the relative tumor residence time and increasing hematopoietic toxicity.
Size, Penetration, and Clearance Rate
Size is one factor that impacts the circulation time of Abs. A full IgG Ab is a large, 150-kDa protein that can remain in circulation for 3–4 weeks while being metabolized slowly by the reticuloendothelial system. A 25-kDa monovalent fragment (i.e., scFv) has a blood clearance time of less than 10 hours with primarily renal excretion in 2–4 hours. Molecules with MWs above ~70 kDa, the glomelular filtration threshold, remain in circulation much longer than smaller, more rapidly eliminated molecules. Smaller molecules tend to exhibit faster uptake, but consequently in lower amounts and with shorter target residence times. Monovalent fragments (i.e. Fab) illustrate this condition. As a result, intermediate-sized minibodies and related constructs are also being pursued to address these limitations.
Solid tumor radiation-absorbed doses are generally far less than the levels that are predicted to result in durable clinical responses. The best uptake is approaching ~ 0.1% injected dose per gram of tumor. This limitation may be rationalized by the inherent barriers of macromolecular entry into solid tumors. Tumor-to-blood ratios as high as 30:1 have been reported at late time points, but therapeutic indices have generally been disappointing due to slow uptake and clearance. Improvements leading to faster target accumulation, complementary rapid clearance, and a more favorable therapeutic index deem well for translation to targeted immunotherapy, especially when coupled with short half-lived nuclides.
To image disease, probes must provide detection specificity, and therefore use of Ab based tumor targeting molecular probes is appropriate. However, the images obtained using Abs have typically been sub-optimal due to long circulation times and concomitant low tumor-to-background ratios. Small-size tumor imaging requires high image contrast. A systemically delivered radiotracer must accumulate at high levels within target sites and clear from the blood in a time frame compatible with the radionuclide's t½.
Increased tumor uptake has been observed for intermediate-sized bivalent Ab formats, such as 75-kDa triabodies and 80-kDa minibodies (scFv-CH3), however, slower blood clearance was evident.64, 65 Faster clearance rates can lead to increased tumor-to-background ratios, enhanced contrast, and improved tumor delineation. For instance, an 123I-anti-CEA minibody allowed imaging of tumors ≥ 1 cm in size by SPECT66, and an 124I-labeled anti-HER2 diabody showed promising results in PET imaging of mouse tumor xenografts.67 Significantly improved blood clearance (from 12 days to 8 hours) was achieved through point mutations in the FcRn receptor of a 105-kDa scFv-Fc molecule, yielding high tumor-to-blood ratios and impressive imaging results.68
Pre-Targeting Approaches
Multistep targeting strategies continue to be investigated and have demonstrated impressive in vitro improvement (factors of 10 or more) over traditional infusion routes.69–71 The biotin-avidin chemical complex may be exploited in multistep approaches.72, 73 For instance, a three-step pre-targeting approach involves biotinylation of the Ab prior to infusion and cell targeting. Injection of avidin thereafter results in exposed avidin sites on the tumor, permitting injection of radiolabeled biotin that either rapidly binds at the tumor complex or concomitantly rapidly clears. Encouraging results have been achieved in pre-clinical settings and, while tumor-to-tissue ratios improved, renal accumulation and toxicity became a potential problem.
NeoRX extensively investigated a three-step strategy of systemic injection of a streptavidin-anti-Ep-CAM Ab conjugate (SA-NR-Lu-10) and radiolabeled biotin with a chase step using either HSA-galactose-biotin73, 74 or biotin-LC-NM-(Gal-NAc)16.75, 76 Despite impressive pre-clinical results, very disappointing results were obtained in a Phase II clinical trial of 90Y-DOTA-biotin pre-targeted by NR-LU-10 Ab/streptavidin in metastatic colon cancer patients.77 The overall response rate was only 8%, and both hematological (8% grade 3 neutropenia, 8% grade 3 thrombocytopenia) and non-hematological (16% grade 3 diarrhea) toxicities were observed.77 The expression of the Ep-CAM receptor in healthy bowel suggests that a poor choice of target receptor, rather than the pre-targeting strategy itself, was the major pitfall generating these negative results.
Despite the lackluster NeoRX results, pre-targeting has produced encouraging results in other cases. Signal amplification by pre-targeting a trivalent, bispecific Ab resulted in a > 40-fold tumor-to-blood ratio increase relative to arcitumomab (99mTc-antiCEA- F(ab’).78 Binding of a divalent hapten labeled with 111In or Gd(III) to scFv receptors allowed for noninvasive assessment by γ or MR imaging of transgenic gene expression.79
Pharmacokinetic behavior
High renal uptake can lead to renal toxicity thereby limiting the amount of administered dose. Similarly, slow clearance from the blood compartment can limit the window during which optimal diagnostic/therapeutic conditions are achieved. It is of critical importance, however, to correctly identify and delineate underlying causes of non-target uptake. The overall charge, hydrophobicity/hydrophilicity, shape, and size can impact pharmacokinetics and clearance as will the presence of endogenous target receptors in non-target organs. Structural modification (e.g., PEGylation) and residue mutation are both useful strategies in reducing chemically- or physically-derived non-target organ uptake of antibodies, but these methods do not reduce uptake due to receptor expression within non-target organs.
In vivo, radioimmunoconjugates accumulate in the liver and kidneys where protein catabolism and elimination occur, and non-specific radioactivity retention results from the transfer of radioactive metal to endogenous metalloproteins. This undesirable behavior can be minimized through various strategies in an effort to increase the tumor-to-normal tissues radiation ratio (i.e., therapeutic index). Cleavable linkers can reduce renal and hepatic doses. Renal retention can also be minimized by the administration of lysine, a cationic amino acid.80
Factors Affecting Tumor Uptake
Studies involving human tumor heterografts in immunosuppressed mice have created unrealistic expectations for RIT. Radionuclide conjugates routinely target established transplants with great efficiency and produce long-term regressions in mice, but such success is rarely observed in cancer patients. Valency, shape, size, isoelectric pH (pI), blood pool concentration, and receptor binding affinity are all parameters of a given Ab that govern the magnitude and depth (penetration) of tumor uptake. Studies with iodine-radiolabeled scFvs demonstrated that a threshold affinity between 10−7 and 10−8 M was required to observe detectable tumor uptake in mice 24 hours post-injection, whereas no gain in tumor accumulation was observed with affinities > 10−9 M.81 Affinities > 10−9 M were detrimental to rapid and uniform tumor penetration due to stable binding at first pass of tumor antigens forming a binding site barrier.81 A 16-amino-acid-long basic cell-penetrating peptide, penetratin, improved solid tumor uptake and distribution when co-injected with an 125I-labeled covalent dimeric scFv.82
Physiological Side Effects
Ideally, a given Ab would exhibit only a single physiological action: binding to its antigen. This is, of course, far from reality. Physiological side effects following Ab infusion are typically less of a concern in the context of radiolabeled diagnostic and RIT Abs since the amounts of protein administered are significantly lower with respect to non-radioactive immunotherapy applications. Nonetheless, unwanted biological effects are real, are often difficult to predict, and must be addressed.
Within 90 minutes following a single Ab infusion, serious physiological complications — including severe pain, extreme swelling, headache, nausea, diarrhea, myalgias, vasodilation, and hypotension — occured in six healthy individuals in TeGenero’s Phase I clinical trial with an unconjugated Ab, anti-CD28 mAb, in 2006.83–85 One individual remained in a coma for three weeks with heart, liver, and renal failure, pneumonia, septicemia, and gangrene. Radiolabeled antibodies can also produce undesirable side effects. A non-oncologic radioimmunodiagnostic agent, 99mTc-fanolesomab (NeutroSpec™), was approved in the U.S. for use in patients with equivocal presentation of appendicitis. However, following serious adverse events, including two fatalities, the agent was withdrawn in late 2005.86
Antibody Production and Commercialization
Antibodies have emerged as an important drug class as underscored by 18 current FDA-approved therapeutic Abs (17 on the market; one withdrawn) across diverse clinical settings.87–93 Among these are 14 unmodified IgG molecules, two radioimmunoconjugates, one Ab-drug conjugate, and one Fab. Cancer therapy represents one major area in which mAbs have been successful, with eight of the 18 being approved for cancer, including treatment of both solid tumors and hematologic cancers (Table 2).94–100 Commercial development of Ab-based pharmaceuticals has become a burgeoning research area.101 Over 400 Abs are currently in clinical trials (www.ClinicalTrials.gov), and sales of recombinant protein therapeutics are projected to reach $15 billion by 2010.102
Three Abs — rituximab (anti-CD20; Rituxan®, marketed as MabThera® outside the U.S.) for treatment of non-Hodgkin’s lymphoma (NHL), trastuzumab (anti-HER2/neu; Herceptin®) for treatment of breast cancer, and bevacizumab (anti-VEGF, Avastin®) for treatment of metastatic colorectal — represented three of Roche’s top five selling pharmaceutical products, comprise half of the six Abs having greater than $1 billion in individual sales in 2005, and had combined sales greater than $6 billion worldwide in 2005. Genentech developed and currently markets these drugs in the U.S.; Roche holds marketing rights for the rest of the world.
Four radiolabeled Abs have been FDA-approved for cancer diagnosis: CEA-Scan® (arcitumomab), Verluma® (nofetumomab merpentan), OncoScint® (satumomab pendetide), and ProstaScint® (capromab pentetide) (Table 2). Arctiumomab, or carcinoembryonic antigen (CEA) scan (Immunomedics; Morris Plains, NJ), is a 99mTc anti-CEA murine F(ab’) approved for SPECT imaging of colorectal cancer patients. Verluma® is a 99mTc Fab fragment approved for identification of advanced-stage small-cell lung cancer (SCLC). Prostascint® (Cytogen Corp.; Princeton, NJ) is an 111In anti-prostate-specific membrane antigen (PSMA) murine IgG currently used for SPECT imaging of post-prostatectomy patients with increasing PSMA. Among these four agents, only ProstaScint® remains available; the others are obsolete.
In addition, two radiolabeled Abs have been FDA-approved for use in nononcologic medical diagnostic imaging: 111In-imiciromab pentetate (MyoScint™, 1996) for myocardial infarctions and 99mTc-fanolesomab (NeutroSpec™, 2004) for equivocal appendicitis. Four additional immunodiagnostic agents have been approved exclusively in Europe: 111In-igovomab (Indimacis 125, 1996) for ovarian adenocarcinoma, 99mTc-murine anti-melanoma fragments (Tecnemab K1, 1996) for cutaneous melanoma lesions, 99mTc-sulesomab (LeukoScan®, 1997) for bone infection and inflammation in osteomyelitis patients, and 99mTc-votumumab (Humaspect, 1998) for colorectal carcinoma.
Regulatory and Financial Barriers
Despite a steady increase in earned revenues and newly approved products, there are concerns about the failure to obtain FDA approval on the part of several well-publicized and promising mAb candidates at the Phase II and Phase III levels. Several likely reasons for negative Phase III trials of molecular-target-based drugs were highlighted in a review by Saijo: (i) poor predictability of pre-clinical models, (ii) low enrichment levels of molecular target within tumors, (iii) molecular target is not essential for tumor survival, (iv) drug produces inadequate antitumor activity, (v) inappropriate clinical decision-making in Phase I/II → Phase III transition, and (vi) inappropriate clinical study design.103 Among these, items (iii) and (iv) are typically not applicable to RIT, as the radionuclide ultimately provides tumor cytotoxicity. The remaining items, however, are quite relevant and represent challenges that must be overcome in order for radiolabeled antibodies to reach their full potential.
Clinical trials involving RIT have demonstrated partial, short-lived responses in some patients with advanced, solid tumors, but most of the significant advances have occurred in the treatment of hematologic neoplasms. The nature of patient population and strict developmental requirements for radiolabeled Abs are complicating factors contributing to a lackluster representation in the clinic. The 10-year absence of any newly approved imaging agents (none since 1996!) is also a sluggish trend that challenges molecular imaging researchers.104, 105
IV. The target antigen and associated tumor(s)
Choice of Target Receptor/Antigen
A variety of promising targets are currently being evaluated in clinical trials at various stages (Table 3). Optimization of an Ab-based pharmaceutical requires several phases of development including choice of target antigen, binding-site selection, choice of effector functions, molecular design, delivery strategy, and administration route. Tumor-associated antigens and receptors present on the tumor cell surface include CD20 (where CD denotes cluster of differentiation), CD22, prostate-specific membrane antigen (PSMA), mucin 1 (MUC1), carcinoembryonic antigen (CEA), pancarcinoma antigen (TAG-72), sialyl Lewis antigen, HER2/neu receptor, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor, and epidermal growth factor receptor (EGFR). In contrast, vascular endothelial growth factor (VEGF) and integrins (e.g., αvβ3, αIIbβ3, and αvβ5) are more abundant on vascular endothelial cells within newly sprouting blood vessels that nourish the nearby tumor during angiogenesis.
Table 3.
Selected Radiolabeled Antibodies Currently Undergoing FDA Clinical Trials for the Detection and Treatment of Cancer (accessed at www.ClinicalTrials.gov.).
Sponsor | Agent/Target | Cancer Indication | Phase |
---|---|---|---|
GlaxoSmithKline | 131I-tositumomab vs. ibritumomab tiuxetan | non-Hodgkin's lymphoma | III |
Radboud University / Immunomedics, Inc. | hMN-14xm734 + 111In-IMP-205 (pretargeted) | colorectal carcinoma | I |
Immunomedics, Inc. | 99mTc-LL2 (LymphoScan) | non-Hodgkin's lymphoma | III |
Immunomedics, Inc. | 90Y-HuPAM4 | pancreatic cancer | I |
Immunomedics, Inc. | 90Y-epratuzumab (hLL2) | non-Hodgkin's lymphoma | II |
Ludwig Institute for Cancer Research | 131I-huA33 (in combination with capecitabine) | metastatic colorectal carcinoma | I |
Ludwig Institute for Cancer Research / Wyeth | 111In-CMD-193 (a humanised mAb linked to the toxin, calicheamicin) | advanced tumors expressing the Lewis-Y antigen | I |
Ludwig Institute for Cancer Research | 111In-cG250 (phase I) and 177LucG250 (phase II), (a chimeric mAb) | advanced renal cell carcinoma | II |
Sidney Kimmel Comprehensive Cancer Center / NCI | 111In-labeled humanized PAM4 IgG | pancreatic cancer | I |
Memorial Sloan-Kettering Cancer Center / NCI | 131I-8H9 | CNS or leptomeningeal cancer | I |
Memorial Sloan-Kettering Cancer Center / NCI | 213Bi-M195 (humanized anti-CD33 mAb) | advanced myeloid cancer | II |
Fred Hutchinson Cancer Research Center | 131I-BC8 | acute myeloid leukemia or myelodysplastic syndromes | II |
Duke University / NCI | 211At-81C6 (antitenascin human/mouse chimeric mAb) | primary or metastatic brain tumors | II |
Weill Medical College of Cornell University / Columbia University | 177Lu-J591 | prostate cancer | II |
NCI | 99mTc-arcitumomab (IMMU-4), comparison study with FDG | colorectal carcinoma | II |
NCI | 131I-TNT-1/B | glioblastoma multiforme | I |
The ideal antigen is readily accessible, highly over-expressed and expressed only within the desired target tissue. One of the limitations of the obsolete 111In-capromab pendetide scan was pooling of the Ab and decreased sensitivity as a result of the epitope’s cytoplasmic location. In the context of RIT, internalization of the receptor after binding carries the radionuclide intracellularly and may lead to infliction of greater damage to nuclear DNA. However, this same action may compromise repeat dosing due to disappearance of the target. Low receptor numbers can yield poor image intensity and limit therapeutic efficacy.
Some targets are less optimal because of shedding or secretion from the cell surface, circulation in the blood, and residence in the interstitial compartment. Freely circulating antigen binds administered Ab, diverting it from cancer cells. Non-Hodgkin’s lymphoma (NHL) idiotype is an example of a shed antigen found within hematologic neoplasms. Many antigens studied extensively as targets for RIT in solid tumors (e.g, CEA and TAG-72) are also secreted, and Abs with these targets will bind circulating target antigen, compromising Ab binding to the tumor. In contrast, CD20 is an excellent target for immunotherapy because it is neither shed from the cell surface nor internalized, and it is expressed by nearly all B-cell tumors.
Antigens found in multiple cancer types
Some target antigens are disease specific, while others are more broadly expressed. Antigens ubiquitous across several tumor types are attractive targets from a commercial viewpoint due to a more populated customer base. Several currently approved immunotherapeutic Abs are targeted towards fairly ubiquitous antigens, many of which are discussed herein, including CEA, TAG-72, HER2/neu, EGFR and vascular endothelial growth factor (VEGF). TAG-72 is expressed on human carcinomas including colorectal, gastric, pancreatic, ovarian, endometrial, breast, non-small cell lung, and prostate. CEA is expressed on colorectal, pancreatic, gastric, non-small cell lung, and breast carcinoma. Because RIT requires differential targeting to minimize hematopoietic toxicity, antigenic distribution would ideally be limited to cancer cells.
A particularly noteworthy example of an Ab directed against a pancarcinoma antigen is nimotuzumab (TheraCIM, YM BioSciences), a humanized mAb directed against a widely expressed transmembrane receptor tyrosine kinase — namely, the EGF receptor (i.e. EGFR, HER-1, or ErbB-1).106 A 188Re-labeled version of the Ab (RadioTheraCIM) showed promising results in a Phase I trial for the radiation treatment of gliomas via an in-dwelling catheter into the post-operative cavity following resection.107 Radioimmunoscintigraphy of nodal metastatic disease using 99mTc-nimotuzumab (DiaCIM) was also performed in a Phase I trial, but little correlation between EGFR expression and positive tumor imaging was observed.108 In addition, unconjugated nimotuzumab (TheraCIM) demonstrated very encouraging results in pediatric pontine (brainstem) and adult gliomas, is undergoing clinical trials in non–small cell lung cancer, and is currently undergoing a Phase II monotherapy trial in Europe in patients with advanced metastatic pancreatic cancer.106 Other potential indications under investigation include nasopharyngeal cancer, head and neck cancer, esophageal cancer, breast cancer, prostate cancer, uterine cervical cancer, and colorectal cancer.106.
Solid Tumors vs. Hematologic Burden
Contrary to clinical responses obtained for hematologic malignancies, solid carcinomas demonstrate an inconsistent response record.109 Investigators are now exploring various enhancement mechanisms designed to increase clinical responses to the levels achieved for B-cell lymphomas. Strategies include increased target tissue localization, decreased normal tissue exposure, and development of more cytotoxic radionuclides and ligand constructs.
Inhomogeneous targeting may be more significant for solid tumors, which are often poorly vascularized110 and have high interstitial pressure due to poor lymphatic drainage, all of which serve to impede entry of macromolecules. Diffusion from blood vessels and tumor penetration are the initial barriers encountered by systemically delivered Abs, and increased interstitial pressure in solid tumors correlates to penetration of a scFv being 6-fold greater than for an IgG.
Higher radioresistance of solid tumors has been proposed as an obstacle to successful RIT, since actual targeting has been shown to be as good, if not better, than that observed in lymphoma. Based on external radiation, solid tumor radiation absorbed dose has been far less than that considered necessary for solid tumor control. Clearly, radioresponse characteristics of indolent B-cell NHL heavily impacted clinical results. Also noteworthy is that the current lymphoma RIT success is as much a feature of the tumor-killing properties of the Ab itself as the radiation-absorbed dose, with no clear relationship between absorbed dose and response.
Following this pathway, other radiolabeled Abs specific to hematopoietic surface antigens are in clinical trials, and one or two additional RIT agents may receive FDA approval in the near term for the treatment of lymphoid malignancies. However, the larger question remains whether RIT will ever become a significant factor in the treatment of the more commonly occurring solid tumors. For several well-vascularized target sites (e.g., breast, ovarian, and brain tumors), intravenous Ab infusion has been the preferred mode of administration. For other regional tumor sites in more enclosed spaces, such as the peritoneum or the CNS, intracompartmental treatments including intraperitoneal (IP), intrathecal (IT), and even intracranial (IC) modes of administration have been employed to increase the accessibility and amount of Ab targeted to tumors. Realistically, adjuvant treatment of compartmental or microscopic residual disease is probably the only attainable indication for solid tumors in the near term.
Clinically significant anti-tumor responses remain quite rare. Efforts to increase administered radiation doses through bone marrow or peripheral stem cell support have not significantly improved solid tumor responses. Combinations of high-dose RIT with chemotherapy have also fallen short of desireable effects. Again, RIT as a stand-alone therapy seems more likely to succeed in a minimal disease burden arena or when used as an adjuvant treatment.
Lymphoma
The development of RIT for lymphoma is more advanced than for any other tumor type. Abs that target specific antigens on B cells include anti-B1 (anti-CD20), LYM-1 (anti- HLA-DR10β), LL2 (anti-CD22), and ibritumomab tiuxetan (anti-CD20). 11, 111
The only two RIT agents approved for commercial use, Bexxar® and Zevalin® both target CD20 and are indicated for treatment of indolent B cell lymphoma and related conditions.11 Both are administered intravenously in a multi-step process over a 1–2 week period. Excellent clinical results are obtained (20–40% complete response rates and 60–80% overall response rates), and toxicity is typically quite mild. Patients with bone marrow involvement by lymphoma (>25%), however, are generally excluded to avoid potential damage to hematopoietic stem cells.
Zevalin® (ibritumomab tiuxetan) was approved in 2002.53 The Zevalin® kit contains a murine mAb and materials for the preparation of both 111In-Zevalin® (for imaging) and 90Y-Zevalin® (for therapy). Zevalin® is co-administered with the therapeutic Ab, Rituxan®. The private insurance industry’s bellwether, Medicare, reimbursed hospitals for Zevalin® treatment after October 2002. Nevertheless, sales have been limited and in 2004 were only $18.7 million as compared to $19.6 million in 2003.
Bexxar®, approved in 2003,112 is an 131I labeled Ab also with limited sales. Bexxar®, originally developed by Corixa, transferred all rights to its partner GlaxoSmithKline in December 2004. Corixa indicated belief in Bexxar®, but stated that commercial acceptance was “too slow” to continue its funding. These problems illustrate that, even with the blessing of FDA approval, there remain obstacles to real financial success.
LL2 has been evaluated for treating NHL. Immunomedics originally tested several candidate murine mAbs labeled with 131I, and ImmuRAIT-LL2 advanced through Phase I/II trials. However, the murine Ab prevented repeat administration, so ImmuRAIT-LL2 was halted in favor of a humanized Ab to obviate immunogenicity. Epratuzumab (hLL2) has been evaluated in Phase I/II studies labeled with 90Y (complexed by DOTA) or 131I with considerable success.113 The 90Y-labeled hLL2 (LymphoCide-Y-90™) was still recruiting in Phase II trials in France and Germany according to ClinicalTrials.gov at the date of submission.114
Immunomedics has also developed 99mTc labeled (Fab’) bectumomab (99mTc-labeled IMMU-LL2, Lymphoscan®), which has shown promise as a pre-RIT imaging probe for B-cell NHL.115, 116 While it excelled at defining small volume, low-grade disease, performance was variable as a purely diagnostic agent. FDG-PET has also been accepted for imaging NHL and has been directly compared to Lymphoscan®.115 Fifteen of the 21 (71%) sites detected by FDG-PET were also positive using bectumomab. In one patient, additional sites were observed by bectumomab that were not delineated by FDG-PET or by CT.115
Lym-1, which targets the HLA-DR10β subunit expressed on most malignant B cells, has been extensively studied by DeNardo and co-workers and has shown efficacy in treating NHL when labeled with 131I or 67Cu.117 Following treatment with 131I-Oncolym®, the overall response rate in 130 indolent and aggressive NHL patients was 26% (five complete remissions and 13 partial remissions). Commercial development of Oncolym® by Peregrine Pharmaceuticals, Inc. was suspended in 2002. The company stated that this was due to the failure to successfully develop the product coupled with the emergence of superior or equivalent products, such as Bexxar®, Zevalin®, and Rituxan®.118
In May 2002, HGS launched Phase I clinical trials on LymphoRad-131, a radio-iodine-labeled (by MDS Nordion) version of B-lymphocyte stimulator (BLyS) intended for treatment of multiple myeloma and NHL. LymphoRad-131 is potentially applicable in the treatment of a broad range of B-cell tumors, including multiple myeloma, large B-cell lymphomas, follicular B-cell lymphomas, chronic lymphocytic leukemia, and Burkitt's lymphoma. The distribution of LymphoRad-131 receptors on multiple myelomas broadens the potential patient population, since CD20 receptors are absent in this condition. In addition, pre-B cells needed to replenish the normal B-cell population lack the LymphoRad-131 receptor, but carry CD20, suggesting that pre-B cells might escape killing by LymphoRad-131. HGS discontinued clinical development of LymphRad-131 in 2005, despite only mild to moderate reversible toxicity and tumor deposited doses up to 3600 cGy being observed in the Phase I trial.119 The anticipated competition from Zevalin® and Bexxar® were likely heavily weighed in this decision.
Colorectal Cancer
Some of the most advanced efforts relate to both imaging and RIT of gastrointestinal disease.120–122 Heavily pursued antigens for colorectal cancer include Ep-CAM, A33, TAG-72, and CEA.
The NR-LU-10 Ab targets the Ep-CAM receptor. Knox and colleagues reported results of a Phase II clinical trial of 90Y-DOTA-biotin pre-targeted by NR-LU-10 Ab/streptavidin in patients with metastatic colon cancer; however, the results were overshadowed by significant bowel toxicity resulting from antigen expression by normal bowel (see section III).77
Iodine-131 has been targeted to the A33 antigen, a transmembrane glycoprotein of the IgG superfamily. In an open-label, dose escalation, biopsy-based Phase I trial, colorectal patients were treated with a combination of 131I-huA33 and 125I-huA33 one week before surgery.123 No dose-limiting toxicity was observed, and excellent tumor uptake was demonstrated.123 Higher doses were administered in a corresponding Phase I dose escalation trial of 131I-huA33 RIT wherein the maximum tolerated dose was determined to be 40 mCi/m² with excellent targeting resulting in four of the 15 patients having stable disease.124
CC49 is a murine Ab that binds to TAG-72. Iodine-131-CC49 and the chimeric version of the Ab have been used to treat patients with colorectal cancer, but failed to produce significant clinical responses.125 Dehalogenation was an issue such that radiolabeling with 90Y was evaluated in a Phase 1 clinical trial, but both tissue biopsies and imaging using the surrogate 111In confirmed high levels of 90Y deposition in the liver.126 In addition, estimates of absorbed hepatic dose equaled or exceeded that which could be achieved in metastatic tumor sites and raised doubts about the potential of 90Y-CC49.126
Goldenberg and colleagues originally used 131I-labeled polyclonal anti-CEA Abs to identify and treat small groups of patients with recurrent CEA-positive adenocarcinomas.127 Goldenberg founded Immunomedics in 1982 with a focus on anti-cancer Ab products. CEA-Scan® is a murine mAb fragment linked to 99mTc indicated with other standard diagnostic modalities for the detection of recurrent and/or metastatic colorectal cancer. In conjunction with CT, the agent provided information about the presence, location, and extent of disease. CEA-Scan® was approved June 26, 1996, however, Immunomedics ceased commercialization during the 2006 fiscal year, consistent with the company's de-emphasis on their diagnostic business.128
Immunomedics completed a series of Phase I/II clinical trials of 90Y-labeled-hMN14 (labetuzumab, CEA-cide®), a humanized radiolabeled Ab targeting CEA, in patients with colorectal (2000–2004)129 and pancreatic (2000–2003)130 cancers. More recently, the company's focus has shifted towards a radiohalogenated version of the same Ab, 131I-labetuzumab, obtaining impressive results in a German Phase II trial in 19 colorectal cancer patients after salvage resection of liver metasases.131 The majority of patients received only a single infusion of 50–60 mCi/m², and a better than predicted outcome was observed: the survival rate was 51% at 5 years, as compared with the expected rate of 28% derived from an analysis of 1,596 patients.131 However, a similar clinical trial in the U.S. using 90Y-labetuzumab was terminated for unspecified reasons, although one might speculate that the choice of 90Y for treating limited residual disease after surgery may have been a factor.132
Another anti-CEA Ab, T84.66, has undergone a Phase I RIT trial in which the 90Y-labeled chimeric Ab was combined with the radiation-enhancing chemotherapy agent, 5-fluorouracil.133 No objective responses were observed; however, more than half of patients shifted from progressive to stable disease.133
Breast Cancer
Breast cancer imaging and therapy is being aggressively studied in a variety of pre-clinical and clinical models. Targets include MUC1, CEA, and L6. Although some clinical responses have been observed, the level of response is again not nearly as compelling as RIT for hematopoietic malignancies. Studies with 99mTc-labeled anti-CEA monoclonal antibodies and with 111In-labeled or radioiodinated anti-mucin antibodies (e.g., HFMG, B72.3, and anti-TF) have demonstrated the ability of radioimmunoscintigraphy in detecting over 80% of breast cancer lesions, but showed lower sensitivity and specificity for accurate staging of the axillae. Non-specific localization of radiolabeled mAbs in tumor-negative nodes, even following lymphoscintigraphy, appears to be the major factor limiting widespread clinical application of radioimmunoscintigraphy in staging newly diagnosed breast cancer patients.
Monoclonal and, more recently, humanized anti-MUC1 Abs, such as BrE-3, have been developed. A Phase I trial was performed to explore the use of 90Y-BrE-3 murine Ab.134 Although responses were observed, an immune response prevented further use of this Ab.134 A humanized version has been evaluated in a clinical trial, and 8 of 17 patients (47%) showed responses despite failing previous conventional therapies.135 Anti-MUC1 Ab, m170, radiolabeled with 90Y and combined with paclitaxel has progressed to dosimetric studies with measurable tumor regression and partial responses.136 A Phase I trial using 90Y-BrE-3 combined with stem cell support gave objective partial responses.137
First believed to be a specific colon cancer marker, CEA was later shown to be expressed in normal tissues and in cancers from other sites, including breast carcinomas.138 NP-4, a murine anti-CEA Ab, labeled with 131I resulted in therapeutic responses in a Phase I/II study. When 57 patients were treated with 131I-NP-4, modest anti-tumor activity was seen in 12 of 35 assessable patients with 1 partial remission, 4 minor/mixed responses and 7 instances of stabilization of progressing disease.139 Preliminary studies with 99mTc-labeled CEA-Scan® appear to indicate a useful role for this agent in distinguishing between benign and malignant breast lesions in patients with indeterminate mammographic findings.140
The L6 cell surface antigen is highly expressed in breast cancer and is related to a number of cell surface proteins with similar predicted membrane topology implicated in cell growth. Yttrium-90-DOTA-peptide-ChL6 resulted in excellent tumor targeting and an effective therapeutic index pre-clinically.141 Combined modality RIT was also performed using the same radioimmunoconjugate combined with an αvβ3-integrin inhibitor142, with paclitaxel143, with an anti-EGFR mAb58, or with a bcl-2 antisense oligonucleotide. Both positive and negative influences on cure rate and toxicity were observed with these various combinations.144
Prostate Cancer
Prostate-specific membrane antigen (PSMA) is perhaps the most highly studied and well-established target for prostate cancer. The most well-known radiolabeled Ab to PSMA is commercially available ProstaScint® (111In-capromab pendetide, Cytogen Corp, Princeton, NJ), which was FDA approved ten years ago for imaging soft-tissue, but not bone sites, of metastatic prostate cancer for presurgical staging or evaluation of PSMA relapse after local therapy.145, 146 Surgical resection of the prostate is not indicated for patients whose disease has spread outside the prostatic bed. For pre-surgical patients with high-risk disease but negative bone, CT, and MRI scans, capromab was able to identify some patients with positive nodes, thereby sparing them unnecessary surgery. No follow-up studies are available that indicate that high-risk patients with a negative capromab scan have a lower failure rate after surgery.
Capromab is severely compromised by its inability to image bone metastases because it is directed toward an intracellular epitope (N-terminus) of the PSMA molecule that is inaccessible to circulating Ab. This is quite an unfortunate scenario, as bone is the first site of metastatic prostate cancer in 72% of patients. Other prostate cancer targets have also been pursued with variable success. No major responses were observed in therapeutic studies targeting TAG-72 in prostate cancer patients.147 More advanced Abs such as J591 target the extracellular domain of PSMA and therefore may provide significant benefits in the imaging of prostate cancer. Promising results have been obtained using 90Y to treat hormone-refractory metastatic prostate cancer.148 Patient recruitment is ongoing for a Phase II trial to study the efficacy of 177Lu-DOTA-J591 in the treatment of metastatic prostate cancer.148 The additive effects of another radioimmunoconjugate, 90Y-DOTA-peptide-ChL6, combined with taxanes yielded a 67% cure rate, whereas no mice were cured with RIT alone or chemotherapy alone.149
Ovarian Cancer
Success for ovarian cancer has been elusive.150 There are no radiolabeled Abs in late stage clinical development for ovarian cancer, although a number are currently in Phase I/II clinical trials including 90Y-HU3S193 at Memorial Sloan-Kettering and 90Y-CC49 at University of Alabama.
Primary and metastatic ovarian tumors have been detected by radioimmunoscintigraphy using two radio-iodinated anti-MUC-1 mAbs, human milk fat globule antigen 1 and 2 (HMFG1 and HMFG2). Y-90-labeled HMFG-1 murine mAb (pemtumomab) has been used to treat patients with advanced ovarian cancer following conventional therapy.151 Encouraging results were obtained in patients with minimal residual disease with 50% complete remission several years post-treatment.151 Following surgery, chemotherapy, and intraperitoneal RIT, 78% of the 21 patients in complete remission survived for longer than 10 years.151 Unfortunately, 90Y-HMFG-1 then failed to demonstrate a therapeutic effect in a multi-institution international randomized concurrently controlled Phase III clinical trial.152
Clinical evaluation of intravenously administered 131I-labeled chimeric mAb MOv18 directed against the folate receptor153 showed that therapeutic doses could be achieved without normal organ toxicity.154 Immuno-specific localization of 90Y-labeled c-MOv18 on FR-expressing tumors has been demonstrated, suggesting that further studies are warranted.155 The same Ab labeled with the α-emitting halogen, 211At, has also been investigated.44
Lung Cancer
Approved in 1996 but presently clinically obsolete, Verluma® is a 99mTc labeled Fab fragment for identifying advanced stage disease in patients with small cell lung cancer (SCLC).156, 157 In a clinical trial involving 89 confirmed SCLC patients, Verluma® accurately determined whether the disease was limited or extensive 82% of the time. However, if the test indicated limited disease, it failed to image tumors in some body organs in ~23% of patients. Additional standard diagnostic tests, such as bone or CT scan, or a bone marrow biopsy are needed to compensate for these false negative readings.
In 2005, a Phase I study of 90Y-CC49 in advanced non-SCLC patients yielded very disappointing results, warranting the development of a humanized version of CC49.158 There were no objective tumor responses, and both immunogenicity and hematologic toxicities were problematic.158
Brain Cancer
Glioma, the most common and lethal form of primary brain tumor, has been treated with cytoreductive surgery, external-beam irradiation, and systemic chemotherapy. Results have been disappointing owing to tumor invasion into functional brain, chemoresistance, and the blood-brain barrier for systemically delivered therapy. Because brain tumors in adults rarely metastasize outside the cranium, locoregional RIT has been evaluated to treat glioblastoma multiforme (GBM), a malignancy with an extremely poor prognosis that generally kills through local invasion and regional metastasis.159
One new delivery strategy and target is tumor necrosis therapy (TNT) via convection-enhanced delivery (CED) of Cotara® (131I-chTNT, Peregrine Pharmaceuticals, Inc., Tustin, CA), an 131I-labeled chimeric mAb that recognizes a universal, intracellular antigen exposed in the necrotic core of malignant solid tumors for the treatment of malignant glioma. Chimeric chTNT was shown to target tumors by binding to DNA exposed in necrotic zones. The clinical experience to date with 131IchTNT was recently reviewed by Shapiro et al.160
Tenascin-C (TN-C) is an extracellular matrix (ECM) glycoprotein expressed ubiquitously in high-grade gliomas, but not in normal brain.161 The murine IgG2b Ab, 81C6, binds to an epitope within the alternatively spliced fibronectin type III region of TN-C. Intratumoral administration of 131I-81C6 has shown promise in a phase I trial.162 In a more recent phase II study at Duke, the efficacy and toxicity of this 131I-81C6 infused directly into the resection cavity (intracavitary injection) was assessed in 33 patients with previously untreated malignant glioma.163 Dosimetry and SPECT imaging were also performed.164 Median survival for all glioma patients was 86.7 weeks, and for GBM patients was 79.4 weeks, exceeding historic controls obtained by treating with conventional radiotherapy and chemotherapy.163 Reversible hematologic toxicity was observed in 27% of patients, and symptomatic neurologic toxicity occurred in 15%, but only a single patient required re-operation for radionecrosis.163
A three-step pre-targeting approach was evaluated in an Italian trial treating 24 patients with recurrent high-grade glioma after a second surgical debulking and implantation of a catheter into the resection cavity.165 Biotinylated anti-TN-C Ab was infused initially, and avidin was administered after 24 hours, followed 18 hours thereafter by 90Y-biotin.165 In a subsequent Phase II study, 37 patients with high-grade glioma, including 20 patients with GBM, were treated similarly.166 Among 12 historical controls with GBM, median survival was 8 months as compared to 33.5 months in the pretargeted RIT group.166
Renal Cancer
Advanced renal cell carcinoma (RCC) resists traditional therapies and is the subject of intense investigation. Metastatic renal cell carcinoma in 33 patients was treated with an 131I-labeled mouse mAb, 131I-G250.167 Antibody immunogenicity (HAMA) within four weeks post-therapy in all patients restricted therapy to a single infusion.167 In external imaging, 131I-labeled mouse mAb G250 showed excellent localization to all tumors that were ≥ 2 cm.167 Tumor reduction was observed in only 2 patients, while 17 patients had stable disease.167 A follow-up Phase I dose escalation trial showed that fractionation did not significantly improve dose limiting hematopoietic toxicity.168
V. Challenges and Prospects
Failure to Launch
The impressive targeting of a broad range of tumors by diverse Abs combined with the relatively good radiosensitivity of certain cancer cells should have encouraged considerable advances in Ab imaging and RIT development. However, despite the significant advances in immunology, including relatively nonimmunogenic chimeric and humanized Abs, and stable radiolabeling with a variety of radionuclides — halides, metals; α-emitters, β-emitters, Auger electron emitters — progress beyond early Phase II RIT studies in solid tumors has been marginal.109, 169 Furthermore, 18F-FDG continues to dominate the molecular imaging domain. Several key challenges must be confronted to increase the presence and success rate of these increasingly important drugs.
Most of the clinical lymphoma histologies that have proven responsive to RIT are also responsive to other new investigational agents, making it difficult for RIT to become established and integrated into standard clinical practice. The competition for inclusion as a standard of care is brisk. As illustrated earlier, financial disincentives are also present that may discourage patient referral by physicians who lack appropriate licensing to inject RIT agents.55
Current Challenges
Researchers in radioimmunodiagnosis and therapy must clearly identify real clinical needs, discover promising new target antigens, and create therapeutic Abs with superior clinical efficacy and safety. The synergy between molecular imaging and targeted radiotherapy will be strengthened by new generations of therapeutic radiopharmaceuticals coupled with advanced imaging systems in the creation personalized dose regimens. Treatment regimes are often lengthy and result in significant side effects, so an early assessment of treatment effectiveness is critical in patient care management.
Detection of simple anatomical changes in tumor size and shape is already feasible. Functional methods of anatomic imaging can assess tumor response before there are structural changes within the tumor. MRI can detect changes in tumor metabolism early during the course of radiation therapy to assess response or radio-resistance. FDG-PET is extremely sensitive to small changes in tumor metabolism; specifically, upregulation of hexokinase in response to the tumor's need for additional energy. This application of PET will no doubt see tremendous growth during the next decade. In 2006, advanced imaging techniques with CT, MR, PET, and SPECT represented 63% of imaging procedures for cancer patients.170 Due to the expanding utility of these applications, the procedure volume for these modalities is predicted to increase 189% by 2016.170
Radionuclide therapy requires injection of higher activities to reach efficient absorbed doses in radio-resistant solid tumors, while limiting exposure of vital organs (e.g., liver, lung, and kidney). Pre-therapeutic dosimetry studies using PET allow accurate quantification of absorbed radiation doses. Multimodal approaches such as PET/CT also show promise to routinely predict and thereafter measure early responses and non-responses. By screening patients for various tumor receptors and antigens, higher therapeutic efficiency is attainable through individualized (i.e. personalized) medicine.
Choosing Better Targets
Perhaps the most prevalent underlying cause of preclinical and clinical failures in both imaging and RIT is a poor choice of target receptor. Several instances of poorly chosen targets have been noted herein, including NeoRX’s streptavidin-anti-Ep-CAM Ab that was plagued by normal bowel receptor expression73–77, the obsolete 111In-capromab pendetide scan whose cytoplasmic epitope in bone metastases is Ab-inaccessible145, 146, and shed/secreted antigens such as NHL idiotype, CEA, and TAG-72. Significant amounts of time, effort, and financial resources have already been futilely exhausted on the pursuit of several targets that were predestined to fail in theory from their inception. Efforts need to be shifted away from such unsuitable targets and redirected toward the development of novel targets having more favorable properties for Ab imaging and RIT.
Although the prospect of developing a new generation of targets might initially seem to be a practically impossible endeavor, much of the groundwork may already be in place. Several potentially successful Ab-based imaging agents are often discarded — like fish back into the sea — simply because they have failed as non-radioactive therapeutic agents. It is a not uncommon for an Ab under pre-clinical evaluation to successfully target a given receptor but still fail to produce a biological therapeutic response. Although ineffective by themselves, such Abs, when coupled with a radionuclide, still hold great potential because specific and exclusive receptor targeting is the only real prerequisite for both molecular imaging and RIT; the Ab is merely a vehicle, while the radionuclide carries the major workload.
Radioimmunotracers vs. FDG-PET
Despite the initial promise of several radioimmunoimaging agents discussed herein, 18FDG is often preferentially used in the clinic owing to convenience, availability, improved accuracy, and lack of immunogenicity. Strengthening this trend is the fact that FDG is approved for reimbursement for treatment-monitoring applications by the Centers for Medicare and Medicaid Services (CMS) under the National Oncologic PET Registry program. Although FDG-PET has progressively asserted itself as the gold standard of imaging for early tumor detection and assessment of treatment response, it is important to consider its mechanism of action. FDG targets upregulation of hexokinase in rapidly growing tumors; as a result, it may fail to target slower growing tumors, and it may also non-specifically accumulate in any lesion of increased metabolism, including inflammatory processes, resulting in a limited diagnostic specificity in some cases.
To illustrate a scenario in which FDG-PET may not be the optimal imaging agent, consider the case of renal cell carcinoma (RCC). FDG has a fairly low sensitivity as a prognostic and diagnostic agent for RCC. At least 25% of metastatic lesions fail to take up FDG.171 In contrast to FDG, tumor targeting by radiolabeled Abs is not governed by a tumor's metabolic status. These considerations warrant the development of immunoPET agents for conditions such as kidney cancer wherein FDG fails to provide useful diagnostic information.
In contrast to the case of RCC, FDG imaging is superior to current Ab-based imaging agents in the clinical diagnosis of colon carcinoma (CC). In CC patients with a rising CEA level but having negative results on conventional imaging studies, an FDG scan is used to determine the likelihood of value from a second-look laparotomy. Clinical investigators comparatively evaluated FDG versus anti-CEA Ab immunoscintigraphy to localize disease before patients underwent second-look laparotomy.172 FDG detected disease in 23 of the patients, while the CEA scan detected disease in only 4.172 FDG successfully predicted unresectable disease in 90% of patients confirmed at laparotomy, but CEA scan failed to predict unresectable disease in any patient.172 Of 16 patients found to have resectable disease at laparotomy, FDG-PET scan predicted it in 81%, but CEA scan predicted it in only 13%.172 These results lead many clinicians to choose FDG-PET over CEA scans as standard practice. It is critical to realize, however, that although these findings raise doubts regarding the efficacy of CEA as a target antigen, one may not infer that FDG is the preferred diagnostic agent in all situations.
Combination Therapies
Despite common perceptions, RIT should not be considered exclusively as a stand-alone therapy. The clinical advantages and increases in efficacy obtained using combination therapies are becoming more evident. Both unconjugated antibodies and radioimmunoconjugates have shown greater effectiveness when used in combination with a broad range of other therapies including chemotherapeutics (e.g. paclitaxel & docetaxel (taxanes), capecitabine), integrin inhibitors, antisense oligonucleotides, and other antibodies. The rationale behind these improvements can encompass simple additive as well as synergist effects, but the notion that tumor receptor expression can significantly vary from patient to patient is sufficient cause to pursue a multi-pronged attack-strategy against tumor burden.
Despite the several instances of favorable results with combination therapies (several have been noted herein58, 123, 137, 142–144, 149), certain limitations severely discourage a smooth translation into the clinic. As mentioned earlier, the combination of RIT with an αvβ3-integrin inhibitor142, paclitaxel143, an anti-EGFR mAb58, or a bcl-2 antisense oligonucleotide144 had a significantly variable impact on cure rate and toxicity. These results emphasize the clinical potential of combined modality RIT, but also highlight the intrinsic complexity of such multi-component systems. This leads to complicated questions that must be systematically answered to satisfy FDA requirements.
Several criteria must be carefully considered, including the likelihood that different patients will respond to different combination therapies due to individual differences in receptor expression, a situation that may be probed by imaging. In additon, the added cost of multiple drug combinations (i.e. ‘cocktails’) can push them financially out of reach by challenging the healthcare system and third-party payers.173 Weighing the benefits of personalized medicine against practical and financial realities is a difficult challenge that looms in the future of RIT.
Regulatory Barriers
A critical factor to consider is the overall cost and time required to obtain approval for new radiopharmaceuticals. The exploratory investigational new drug (xIND) in the U.S. is intended to decrease the barriers to execute preliminary toxicology-pathology studies. Despite the xIND pathway, study completion times and pathological tissue examination costs continue to rise, and safety concerns contribute to this effect. Nevertheless, the increased use of radiotracers can provide a better understanding of the pharmacologic effect of the parent compound, leading to faster approval processes for first in man studies and, eventually, full clinical approval.
Economic Barriers: Supply and Demand
In the United States, kidney cancer accounts for about 3% of all cancers, with approximately 12,000 deaths each year. With early diagnosis, the survival rate for RCC patients ranges from 79 to 100 percent. As highlighted by Choyke174, however, it is unreasonable to expect the development of new molecular imaging methods that will be able to identify histologic subtypes of RCC in the near term. With only 35,000 new cases of RCC per year in the US and 700,000 worldwide, the commercialization economics of such agents is unfavorable; insufficient numbers of cases exist to justify the required monetary investment associated with the development of a stand-alone diagnostic agent.
For comparison, over 20 million Americans alone take the cholesterol-lowering agent Lipitor® (atorvastatin; Pfizer, New York, NY) every day, and annual worldwide sales are over $12 billion. Comparison to such blockbuster drugs places profits derived from stand-alone diagnostic imaging agents at a severe competitive disadvantage. Molecular imaging agents that identify targets amenable to specific molecular therapies hold more promise. Like their matched treatments, these agents span more than one type of cancer and therefore offer a larger, more viable market for drug developers. Examples include the angiogenic inhibitor and mAb against VEGF, Avastin® (bevacizumab), the mAb against HER2/neu receptors, Herceptin® (trastuzumab; Genentech) and the mAb against HER1 receptors, cetuximab (Erbitux®; ImClone Systems, New York, NY).
Concluding Remarks
The multidisciplinary fields of Ab-based molecular imaging and RIT have yet to reach their full potentials in both pre-clinical and clinical domains. In contrast to the body of impressive pre-clinical data derived from murine models, the limited clinical response rate in RIT has been quite disappointing. This trend extends across several tumor types, antigens, delivery platforms, and radiochemical formats. The only exceptions, hematopoietic neoplasms, are uniquely susceptible to RIT due to high radiosensitivity combined with the immunobiological response to Ab binding. Furthermore, radiolabeled antibodies possess an overwhelming potential in molecular target-based imaging for clinical diagnosis and therapy monitoring, yet FDG continues to be the agent of choice.
Possible solutions to the current problems at hand involve selection of better and new target antigens, humanized or chimeric Abs or fragments, and wiser selection of radionuclides and delivery mechanisms. Molecular imaging will play a pivotal role in shaping the future of targeted RIT, as will advances in imaging equipment and data reconstruction. The creation of personalized treatments and dose regimens will allow therapies to be specifically tailored to match the unique receptor expression of a given patient's tumor burden. In the context of RIT, post-surgical adjuvant treatment of residual disease appears to be the most reasonable short-term clinical goal wherein significant success might be realized. In the future, however, additional technical advances may also help penetrate the more difficult barriers impeding treatment and control of gross disease deposits associated with solid tumors. Finally, the integration of RIT from a stand-alone therapy into a component of standard combination therapies taking advantage of synergistic activities should also be pursued.
Acknowledgement
This research was supported [in part] by the Intramural Research Program of the NIH, National Cancer Institute, Center for Cancer Research. We would also like to acknowledge salient discussions with Otis B. Driftwood and suggestions from Quincy Adams Wagstaff.
Abbreviations
- Ab
antibody
- FDA
U.S. Food and Drug Administration
- RIT
radioimmunotherapy
- FDG
fluorodeoxyglucose
- PET
positron emission tomography
- t½
half-life
- CEA
carcinoembryonic antigen
- NHL
non-Hodgkin's lymphoma
- CD
cluster of differentiation
- mAb
monoclonal antibody
- γ
gamma ray
- SPECT
single-photon emission computerized tomography
- MRI
magnetic resonance imaging
- CT
computed tomography
- eV
electron volts
- β or β−
beta particle (negatron)
- β+
positron
- Gy
gray (absorption of 1 joule of radiation energy by 1 kilogram of matter)
- LET
linear energy transfer
- DNA
deoxyribonucleic acid
- α
alpha particle (helium nucleus)
- RBE
relative biologic effectiveness
- MDS
medical device sterilization
- NBC
National Broadcasting Company
- DTPA
diethylenetriaminepentaacetic acid
- DOTA
1,4,7,20-tetraazacyclododecane N,N,′,N″,N‴-tetraacetic acid
- Ig
immunoglobulin
- CHX-DTPA
2-(p-isothiocyanato-benzyl)-cyclohexyl-diethylenetriaminepentaacetic acid
- TETA
1,4,8,11-tetraacetic acid
- SOD
superoxide dismutase
- CB-TE2A
4,11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecane
- cGMP
current Good Manufacturing Practices
- MeO-DOTA
methoxy-DOTA
- mTCR
monoclonal T cell receptor
- EDTMP
ethylenediamine-tetra (methylene phosphonic acid)
- DOTMP
1,4,7,10-tetraazacyclododecane–1,4,7,10-tetramethylene-phosphonate
- HGS
Human Genome Sciences
- MX-DTPA
combined 1-p-isothiocyanatobenzyl 3-methyl- and 1-p-isothiocyanatobenzyl 4-methyldiethylenetriamine pentaacetic acid
- HAMA
human antimouse antibody
- CDR
complementarity determining region
- scFv
single-chain variable fragment
- Da
Dalton
- Fab
antigen-binding fragment
- FcRn
neonatal Fc receptor
- SA
streptavidin
- Ep-CAM
epithelial cellular adhesion molecule
- HSA
human serum albumin
- Gal-NAc
N-acetyl galactosamine
- PEG
polyethylene glycol
- HER
human epidermal growth factor receptor
- VEGF
vascular endothelial growth factor
- PSMA
prostate-specific membrane antigen
- MUC
mucin
- TAG-72
pancarcinoma antigen
- TRAIL
tumor necrosis factor-related apoptosis-inducing ligand
- EGFR
epidermal growth factor receptor
- ErbB
epidermal growth factor receptor
- IP
intraperitoneal
- IT
intrathecal
- IC
intracranial
- BLyS
B-lymphocyte stimulator
- Lym
lymphoma
- CC
colorectal carcinoma
- HMFG
human milk fat glubule antigen
- FR
folate receptor
- SCLC
small cell lung cancer
- GBM
glioblastoma multiforme
- TNT
tumor necrosis therapy
- CED
convection-enhanced delivery
- RCC
renal cell carcinoma
- ECM
extracellular matrix
- xIND
exploratory investigational new drug
- CMS
Centers for Medicare and Medicaid Services
Footnotes
Portions of this article were highlighted during a presentation at the Workshop on Molecular Imaging: After Bench to Bedside: Impact on Clinical Outcome Feb. 23–25, 2007.
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.
References
- 1.Milenic DE, Brady ED, Brechbiel MW. Antibody-targeted radiation cancer therapy. Nat Rev Drug Disc. 2004;3:488–496. doi: 10.1038/nrd1413. [DOI] [PubMed] [Google Scholar]
- 2.Govindan SV, Griffiths GL, Hansen HJ, Horak ID, Goldenberg DM. Cancer therapy with radiolabeled and drug/toxin-conjugated antibodies. Tech Cancer Res Treat. 2005;4(4):375–391. doi: 10.1177/153303460500400406. [DOI] [PubMed] [Google Scholar]
- 3.Ng DCE. Radioimmunotherapy: a brief overview. Biomed Imaging Interv J. 2006;2(3):e23. doi: 10.2349/biij.2.3.e23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Pohlman B, Sweetenham J, Macklis RM. Review of clinical radioimmunotherapy. Expert Rev Anticancer Ther. 2006;6(3):445–461. doi: 10.1586/14737140.6.3.445. [DOI] [PubMed] [Google Scholar]
- 5.Ehrlich P. The collected papers of Paul Ehrlich. Vol. 3 London: Pergamon; 1960. [Google Scholar]
- 6.Pressman D. Tissue localizing antibodies. Ann N Y Acad Sci. 1955;59(3):376–380. doi: 10.1111/j.1749-6632.1955.tb45950.x. [DOI] [PubMed] [Google Scholar]
- 7.Mach JP, Carrel S, Merenda C, Sordat B, Cerottini JC. In vivo localization of radiolabeled antibodies to carcinoembryonic antigen in human colon carcinoma grafted into nude mice. Nature. 1974;248(450):704–706. doi: 10.1038/248704a0. [DOI] [PubMed] [Google Scholar]
- 8.Goldenberg DM, Preston DF, Primus FJ, Hansen HJ. Photoscan localization of GW-39 tumors in hamsters using radiolabeled anticarcinoembryonic antigen immunoglobulin G. Cancer Res. 1974;34(1):1–9. [PubMed] [Google Scholar]
- 9.Kohler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975;256:495–497. doi: 10.1038/256495a0. [DOI] [PubMed] [Google Scholar]
- 10.Bierman PJ, Vose JM, Leichner PK, Quadri SM, Armitage JO, Klein JL, Abrams RA, Dicke KA, Vriesendorp HM. Yttrium 90-labeled antiferritin followed by high-dose chemotherapy and autologous bone marrow transplantation for poor-prognosis Hodgkin's disease. J Clin Onc. 1993;11(4):698–703. doi: 10.1200/JCO.1993.11.4.698. [DOI] [PubMed] [Google Scholar]
- 11.Sharkey RM, Burton J, Goldenberg DM. Radioimmunotherapy of non-Hodgkin's lymphoma: a critical appraisal. Expert Rev Clin Immunol. 2005;1(1):47–62. doi: 10.1586/1744666X.1.1.47. [DOI] [PubMed] [Google Scholar]
- 12.Jaffer FA, Weissleder R. Molecular imaging in the clinical arena. JAMA. 2005;293:855–862. doi: 10.1001/jama.293.7.855. [DOI] [PubMed] [Google Scholar]
- 13.Kelloff GJ, Krohn KA, Larson SM, Weissleder R, Mankoff DA, Hoffman JM, Link JM, Guyton KZ, Eckelman WC, Scher HI, O'Shaughnessy J, Cheson BD, Sigman CC, Tatum JL, Mills GQ, Sullivan DC, Woodcock J. The progress and promise of molecular imaging probes in oncologic drug development. Clin Cancer Res. 2005;11(22):7967–7985. doi: 10.1158/1078-0432.CCR-05-1302. [DOI] [PubMed] [Google Scholar]
- 14.Barbet J, Chatal JF, Gauche F, Martino J. Which radionuclides will nuclear oncology need tomorrow? Eur J Nucl Med Mol Imaging. 2006;33(6):627–630. doi: 10.1007/s00259-006-0116-4. [DOI] [PubMed] [Google Scholar]
- 15.Carlsson J, Aronsson EF, Hietala SO, Stigbrand T, Tennvall J. Tumour therapy with radionuclides: assessment of progress and problems. Radiother Oncol. 2003;66:107–117. doi: 10.1016/s0167-8140(02)00374-2. [DOI] [PubMed] [Google Scholar]
- 16.Anderson CJ, Connett JM, Schwarz SW, Rocque PA, Guo LW, Philpott GW, Zinn KR, Meares CF, Welch MJ. Copper-64-labeled antibodies for PET imaging. J Nucl Med. 1992;33:1685–1691. [PubMed] [Google Scholar]
- 17.Anderson CJ, Schwarz SW, Connett JM, Cutler D, Guo LW, Germain CJ, Philpott GW, Zinn KR, Greiner DP, Meares CF, Welch MJ. Preparation, biodistribution and dosimetry of 64Cu-labeled anti-colorectal carcinoma monoclonal antibody (MAb) fragments 1A3 F(ab')2. J Nucl Med. 1995;36(5):850–858. [PubMed] [Google Scholar]
- 18.Anderson CJ, Schwarz SW, Connett JM, Rocque PA, Guo LW, Zinn KR, Philpott GW, Welch MJ. Optimization of the preparation of 64Cu-labeled monoclonal antibody (MAb) fragments [1A3-F(ab')2] for improved biodistribution. J Nucl Med. 1993;34:28P. [Google Scholar]
- 19.Verel I, Visser GWM, Boellaard R, Boerman OC, van Eerden J, Snow GB, Lammertsma AA, van Dongen GAMS. Quantitative 89Zr immuno-PET for in vivo scouting of 90Y-labeled monoclonal antibodies in xenograft-bearing nude mice. J Nucl Med. 2004;44(10):1663–1670. [PubMed] [Google Scholar]
- 20.Verel I, Visser GWM, Boellaard R, Stigter M, Snow GB, van Dongen GAMS. 89Zr Immuno-PET: comprehensive procedures for the production of 89Zr-labeled monoclonal antibodies. J Nucl Med. 2004;44(8):1271–1281. [PubMed] [Google Scholar]
- 21.Verel I, Visser GWM, Boerman OC, van Eerden J, Finn R, Boellaard R, Vosjan JWD, Stigter M, Snow GB, van Dongen GAMS. Long-lived positron emitters 89Zr and 124I for scouting of therapeutic radioimmunoconjugates with PET. Cancer Biother Radiopharm. 2004;18(4):655–660. doi: 10.1089/108497803322287745. [DOI] [PubMed] [Google Scholar]
- 22.Verel I, Visser GWM, van Dongen GA. The promise of immunoPET in radioimmunotherapy. J Nucl Med. 2005;46:164S–171S. [PubMed] [Google Scholar]
- 23.Borjesson PKE, Jauw YWS, Boellaard R. Performance of immunoPET with 89Zr-labeled chimeric monoclonal antibody U36 in the detection of lymph node metastases in head and neck cancer patients. Clin Cancer Res. 2006;12:2133–2140. doi: 10.1158/1078-0432.CCR-05-2137. [DOI] [PubMed] [Google Scholar]
- 24.Leichner PK, Kwok CS. Tumor dosimetry in radioimmunotherapy: Methods of calculation for beta particles. Med Phys. 1993;20(2 Pt 2):529–534. doi: 10.1118/1.597048. [DOI] [PubMed] [Google Scholar]
- 25.O'Donoghue JA, Bardies M, Wheldon TE. Relationships between tumor size and curability for uniformly targeted therapy with beta-emitting radionuclides. J Nucl Med. 1995;36(10):1902–1909. [PubMed] [Google Scholar]
- 26.Anderson CJ, Connett JM, Baumann ML, Schwarz SW, Zinn KR, Philpott GW, Welch MJ. Comparison of 67Cu and 64Cu as potential radionuclides for radiotherapy. J Nucl Med. 1993;34:134P. [Google Scholar]
- 27.Apelgot S, Coppey J, Gaudemer A, Grisvard J, Guille E, Sasaki I, Sissoeff I. Similar lethal effect in mammalian cells for two radioisotopes of copper with different decay schemes, 64Cu and 67Cu. Int J Radiat Biol. 1989;55:365–384. doi: 10.1080/09553008914550421. [DOI] [PubMed] [Google Scholar]
- 28.Behr TM, Behe M, Stabin MG, Wehrmann E, Apostolidis C, Molinet R, Strutz F, Fayyazi A, Wieland E, Gratz S, Koch L, Goldenberg DM, Becker W. High-linear energy transfer (LET) α versus Low-LET β emitters in radioimmunotherapy of solid tumors: therapeutic efficacy and dose-limiting toxicity of 213Bi- versus 90Y-labeled CO17-1A Fab' fragments in a human colonic cancer model. Cancer Res. 1999;59:2635–2643. [PubMed] [Google Scholar]
- 29.Allen BJ, Raja C, Rizvi S, Li Y, Tsuji J, Zhang D, Song E, Fa Qu C, Kearsley J, Graham P, Thompson J. Targeted alpha therapy for cancer. Phys Med Biol. 2004;49:3703–3712. doi: 10.1088/0031-9155/49/16/016. [DOI] [PubMed] [Google Scholar]
- 30.Fang N. Research on 212Pb- and 212Bi- radiopharmaceuticals for therapy. J Isot. 1997;10(2):114–118. [Google Scholar]
- 31.Henriksen G, Schoultz BW, Hoff P, Larsen RH. Potential in vivo generator for alpha-particle therapy with 212Bi: presentation of a system to minimize escape of daughter nuclide after decay of 212Pb to 212Bi. Breast Cancer Res Treat. 2003;2003(91):109–113. [Google Scholar]
- 32.Rotmensch J, Atcher RW, Schlenker R, Hines J, Grdina D, Block BS, Press MF, Herbst AL, Weichselbaum RR. The effect of the α-emitting radionuclide 212Pb on human ovarian carcinoma: a potential new form of therapy. Gyn. Oncol. 1989;32:236–239. doi: 10.1016/s0090-8258(89)80040-x. [DOI] [PubMed] [Google Scholar]
- 33.Ruble G, Wu C, Squire RA, Gansow OA, Strand M. The use of 212Pb-labeled monoclonal antibody in the treatment of murine erthyroleukemia. Int J Radiat Oncol Biol Phys. 1996;34:609–616. doi: 10.1016/0360-3016(95)02119-1. [DOI] [PubMed] [Google Scholar]
- 34.Su FM, Beaumier P, Axworthy D, Atcher R, Fritzberg A. Pre-targeted radioimmunotherapy in tumored mice using an in vivo212Pb/212Bi generator. Nucl Med Biol. 2005;32:741–747. doi: 10.1016/j.nucmedbio.2005.06.009. [DOI] [PubMed] [Google Scholar]
- 35.Apostolidis C, Carlos-Marques R, Janssens W, Molinet R, Nikula T, Ouadi A. Production of carrier free 225Ac/213Bi from 229Th for alpha-immunotherapy. J Labelled Cpd Radiopharm. 2001;44(1):5806–5808. [Google Scholar]
- 36.Apostolidis C, Carlos-Marques R, Janssens W, Nikula T, Ouadi A. Cancer treatment using 213Bi and 225Ac in radioimmunotherapy. Nuclear News. 2001 December;:29–33. [Google Scholar]
- 37.Apostolidis C, Molinet R, Rasmussen G, Morgenstern A. Production of 225Ac from 229Th for targeted α therapy. Anal Chem. 2005;77(19):6288–6291. doi: 10.1021/ac0580114. [DOI] [PubMed] [Google Scholar]
- 38.Ballangrud AM, Yang WH, Charlton DE, McDevitt MR, Hamacher KA, Panageas KS, Ma D, Bander HN, Scheinberg DA, Sgouros G. Response of LNCaP spheroids after treatment with an α-particle emitter (213Bi)- labeled anti-prostate-specific membrane antigen antibody (J591) Cancer Res. 2001;61:2008–2014. [PubMed] [Google Scholar]
- 39.Ballangrud AM, Yang WH, Palm S, Enmon R, Borchardt PE, Pellegrini VA, McDevitt MR, Scheinberg DA, Sgouros G. Alpha-particle emitting atomic generator 225Ac-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]
- 40.Boll RA, Malkemus D, Mirzadeh S. Production of 225Ac for alpha particle mediated radioimmunotherapy. Appl Radiat Isot. 2005;62:667–679. doi: 10.1016/j.apradiso.2004.12.003. [DOI] [PubMed] [Google Scholar]
- 41.Allen BJ, Rizvi S, Li Y, Tian Z, Ranson M. In vitro and pre-clinical targeted alpha therapy for melanoma, breast, prostate and colorectal cancers. Crit Rev Oncol Hematol. 2001;39:139–146. doi: 10.1016/s1040-8428(01)00113-5. [DOI] [PubMed] [Google Scholar]
- 42.Allen BJ, Tian Z, Rizvi SMA, Li Y, Ranson M. Pre-clinical studies of targeted a therapy for breast cancer using 213Bi-labeled-plasminogen activator inhibitor type 2. Br J Cancer. 2003;88(6):944–950. doi: 10.1038/sj.bjc.6600838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Aurlien E, Larsen RH, Kvalheim G, Bruland OS. Demonstration of highly specific toxicity of the α-emitting radioimmunoconjugate 211At-rituximab against non-Hodgkin's lymphoma cells. Br J Cancer. 2000;83(10):1375–1379. doi: 10.1054/bjoc.2000.1453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Andersson H, Lindegren S, Back T, Jacobsson L, Leser G, Hovath G. Radioimmunotherapy of nude mice with intraperitoneally growing ovarian cancer xenograft utilizing 211At-labeled monoclonal antibody MOv18. Anticancer Res. 2001;20:459–462. [PubMed] [Google Scholar]
- 45.Sundberg AL, Almqvist Y, Orlova A, Blomquist E, Jensen HJ, Gedda L, Tolmachev V, Carlsson J. Combined effect of gefitinib ('Iressa', ZD1839) and targeted radiotherapy with 211At-EGF. Eur J Nucl Med Mol Imaging. 2003;30:1348–1356. doi: 10.1007/s00259-003-1308-9. [DOI] [PubMed] [Google Scholar]
- 46.Allen BJ, Blagojevic N. Alpha- and beta-emitting radiolanthanides in targeted cancer therapy: The potential role of 149Tb. Nucl Med Biol. 1996;17:40–47. doi: 10.1097/00006231-199601000-00008. [DOI] [PubMed] [Google Scholar]
- 47.Aurlien E, Kvinnsland Y, Larsen RH, Bruland OS. Radiation doses to non-Hodgkin's lymphoma cells and normal bone marrow exposed in vitro. Comparison of an α-emitting radioimmunoconjugate and external γ-irradiation. Int J Radiat Biol. 2002;78(2):1333–1142. doi: 10.1080/09553000110094788. [DOI] [PubMed] [Google Scholar]
- 48.Behr TM, Sgouros G, Stabin MG, Behe M, Angerstein C, Blumenthal RD, Apostolidis C, Molinet R, Sharkey RM, Koch L, Goldenberg DM, Becker W. Studies on the red marrow dosimetry in radioimmunotherapy: an experimental investigation of factors influencing the radiation-induced myelotoxicity in therapy with α-, Auger/conversion electron-, or α-Emitters. Clin Cancer Res. 1999;5:3031–3043. [PubMed] [Google Scholar]
- 49.Boswell CA, Brechbiel MW. Auger electrons: lethal, low energy, and coming soon to a tumor cell nucleus near you. J Nucl Med. 2005;46(12):1946–1947. [PubMed] [Google Scholar]
- 50.Behr TM, Behe M, Lohr M, Sgouros G, Angerstein C, Wehrmann E, Nebendahl K, Becker W. Therapeutic advantages of Auger electron-over β- emitting radiometals or radioiodine when conjugates to internalizing antibodies. Eur J Nucl Med. 2000;27(7):753–765. doi: 10.1007/s002590000272. [DOI] [PubMed] [Google Scholar]
- 51.Silberstein EB. Radionuclides and radiopharmaceuticals for 2005. J Nucl Med. 2005;46(5):13N–26N. [Google Scholar]
- 52.Brass JM, Krummen K, Moll-Kaufmann C. Quality assurance after process changes of the production of a therapeutic antibody. Pharmaceutica Acta Helvetiae. 1996;71:395–403. doi: 10.1016/s0031-6865(96)00051-9. [DOI] [PubMed] [Google Scholar]
- 53.Gordon LI, Witzig T, Molina A, Czuczman M, Emmanouilides C, Joyce R, Vo K, Theuer C, Pohlman B, Bartlett N, Wiseman G, Darif M, White C. Yttrium 90-labeled ibritumomab tiuxetan radioimmunotherapy produces high response rates and durable remissions in patients with previously treated B-cell lymphoma. Clin Lymphoma. 2004;5(2):98–101. doi: 10.3816/clm.2004.n.015. [DOI] [PubMed] [Google Scholar]
- 54.Ghielmini M, Schmitz SF, Cogliatti SB, Pichert G, Hummerjohann J, Waltzer U, Fey MF, Betticher DC, Martinelli G, Peccatori F, Hess U, Zucca E, Stupp R, Kovacsovics T, Helg C, Lohri A, Bargetzi M, Vorobiof D, Cerny T. Prolonged treatment with rituximab in patients with follicular lymphoma significantly increases event-free survival and response duration compared with the standard weekly × 4 schedule. Blood. 2004;15:4416–4423. doi: 10.1182/blood-2003-10-3411. [DOI] [PubMed] [Google Scholar]
- 55.Ellis R. Cancer docs profit from chemotherapy drugs. Accessed Feb. 2007 at http://www.msnbc.msn.com/id/14944098/
- 56.Kaden TA. Labelling monoclonal antibodies with macrocyclic radiometal complexes. A challenge for coordination chemists. Dalton Trans. 2006:3617–3623. doi: 10.1039/b606410h. [DOI] [PubMed] [Google Scholar]
- 57.Clifford T, Boswell CA, Biddlecombe GB, Lewis JS, Brechbiel MW. Small animal PET/CT imaging using 86Y-CHX-A"-octreotide: validation of a novel CHX-A" derivative suitable for peptide conjugation. J Med Chem. 2006;49(14):4297–4304. doi: 10.1021/jm060317v. [DOI] [PubMed] [Google Scholar]
- 58.Lee FT, Mountain AJ, Kelly MP, Hall C, Rigopoulus A, Johns TG, Smyth FE, Brechbiel MW, Nice EC, Burgess AW, Scott AM. Enhanced efficacy of radioimmunotherapy with 90Y-CHX-A"-DTPA-hu3S193 by inhibition of epidermal growth factor receptor (EGFR) signaling with EGFR tyrosine kinase inhibitor AG1478. Clin Cancer Res. 2005;11:7080s–7086s. doi: 10.1158/1078-0432.CCR-1004-0019. [DOI] [PubMed] [Google Scholar]
- 59.Vandenbulcke K, De Vos F, Offner F, Philippe J, Apostolidis C, Molinet R, Nikula TK, Bacher K, de Gelder V, Vral A, Lahorte C, Thierens H, Dierckx RA, Slegers G. In vitro evaluation of 213Bi-rituximab versus external gamma irradiation for the treatment of B-CLL patients: relative biological efficacy with respect to apoptosis induction and chromosomal damage. Eur J Nucl Med Mol Imaging. 2003;30(10):1357–1364. doi: 10.1007/s00259-003-1228-8. [DOI] [PubMed] [Google Scholar]
- 60.Boswell CA, Sun X, Niu W, Weisman GR, Wong EH, Rheingold AL, Anderson CJ. Comparative in vivo stability of 64Cu-labeled cross-briged and conventional tetraazamacrocyclic complexes. J Med Chem. 2004;47(6):1465–1474. doi: 10.1021/jm030383m. [DOI] [PubMed] [Google Scholar]
- 61.Wadas T, Anderson CJ. Radiolabeling of TETA- and CB-TE2A-conjugated peptides with 64Cu. Nature Prot. 2007;1(6):3062–3068. doi: 10.1038/nprot.2006.431. [DOI] [PubMed] [Google Scholar]
- 62.The Dow Chemical Company. Accessed Feb. 2007 at http://pharma.dow.com/chelamed/index.htm.
- 63.Baker M. Upping the ante on antibodies. Nature Biotechnol. 2005;23(9):1065–1072. doi: 10.1038/nbt0905-1065. [DOI] [PubMed] [Google Scholar]
- 64.Todorovska A, Roovers RC, Dolezal O, Kortt AA, Hoogenboom HR, Hudson PJ. Design and application of diabodies, triabodies and tetrabodies for cancer targeting. J Immunol Methods. 2001;248(1–2):47–66. doi: 10.1016/s0022-1759(00)00342-2. [DOI] [PubMed] [Google Scholar]
- 65.Wu AM, Yasaki PJ, Tsai SW, Nguyen K, Anderson AL, McCarthy DW, Welch MJ, Shively JE, Williams LE, Raubitschek AA, Wong JYC, Toyokuni T, Phelps ME, Gambhir SS. High-resolution microPET imaging of carcinoemryonic antigen-positive zenografts by using a 64Cu-labeled engineered antibody fragment. Proc Natl Acad Sci USA. 2000;97(15):8495–8500. doi: 10.1073/pnas.150228297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Wong JYC, Chu DZ, Williams LE, Yamauchi DM, Ikle DN, Kwok CS, Liu A, Wilczynski S, Colcher D, Yazaki PJ, Shively JE, Wu AM, Raubitschek AA. Pilot trial evaluating an 123I-labeled 80-kilodalton engineered anticarcinoembryonic antigen antibody fragment (cT84.66 minibody) in patients with colorectal cancer. Clin Cancer Res. 2004;10:5014–5021. doi: 10.1158/1078-0432.CCR-03-0576. [DOI] [PubMed] [Google Scholar]
- 67.Robinson MK, Doss M, Shaller C, Narayanan D, Marks JD, Adler LP, Gonzalez Trotter DE, Adams GP. Quantitative immuno-positron emission tomography imaging of HER2-positive tumor xenograft with an 124I labeled anti- HER2 diabody. Cancer Res. 2005;65(4):1471–1478. doi: 10.1158/0008-5472.CAN-04-2008. [DOI] [PubMed] [Google Scholar]
- 68.Kenanova V, Olafsen T, Crow DM, Sundaresan G, Subbarayan M, Carter NH, Ikle DN, Yazaki PJ, Chatziioannou AF, Gambhir SS, Williams LE, Shively JE, Colcher D, Raubitschek AA, Wu AM. Tailoring the pharmacokinetics and positron emission tomography imaging properties of anticarcinoembryonic antigen single chain Fv-Fc antibody fragments. Cancer Res. 2005;65:622–631. [PMC free article] [PubMed] [Google Scholar]
- 69.Barbet J, Kraeber-Bodere F, Vuillez JP, Gautherot E, Rouvier E, Chatal JF. Pre-targeting with the affinity enhancement system for radioimmunotherapy. Cancer Biother Radiopharm. 1999;14(3):153–165. doi: 10.1089/cbr.1999.14.153. [DOI] [PubMed] [Google Scholar]
- 70.Goldenberg DM, Sharkey RM, Paganelli G, Barbet J, Chatal JF. Antibody pre-targeting advances cancer radioimmunodetection and radioimmunotherapy. J Clin Oncol. 2006;24:823–834. doi: 10.1200/JCO.2005.03.8471. [DOI] [PubMed] [Google Scholar]
- 71.Gruaz-Guyon A, Raguin O, Barbet J. Recent advances in pre-targeted radioimmunotherapy. Curr Med Chem. 2005;12:319–338. doi: 10.2174/0929867053363225. [DOI] [PubMed] [Google Scholar]
- 72.Breitz HB, Fisher DR, Goris ML, Knox S, Ratliff B, Murtha AD, Weiden PL. Radiation absorbed dose estimation for 90Y-DOTA-biotin with pretargeted NR-LU-10/streptavidin. Cancer Biother Radiopharm. 1999;14(5):381–395. doi: 10.1089/cbr.1999.14.381. [DOI] [PubMed] [Google Scholar]
- 73.Breitz HB, Weiden PL, Beaumier PL, Axworthy DB, Seiler C, Su FM, Graves S, Bryan K, Reno JM. Clinical optimization of pre-targeted radioimmunotherapy with antibody-streptavidin conjugate and 90Y-DOTA-biotin. J Nucl Med. 2000;41:131–140. [PubMed] [Google Scholar]
- 74.Axworthy DB, Reno JM, Hyalarides MD, Mallett RW, Theodore LJ, Gustavson LM, Su FM, Hobson LJ, Beaumier PA, Fritzberg AR. Cure of human carcinoma xenografts by a single dose of pre-targeted 90Y with negligible toxicity. Proc Natl Acad Sci USA. 2000;97:1802–1807. doi: 10.1073/pnas.97.4.1802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Press OW, Corcoran M, Subbiah K, Hamlin DK, Wilbur DS, Johnson T, Theodore L, Yau E, Mallett R, Meyer DL, Axworthy D. A comparative evaluation of conventional and pre-targeted radioimmunotherapy of CD20-expressing lymphomas xenografts. Blood. 2001;98(15):2535–2543. doi: 10.1182/blood.v98.8.2535. [DOI] [PubMed] [Google Scholar]
- 76.Weiden PL, Breitz HB. Pre-targeted radioimmunotherapy (PRIT) for treatment of non-Hodgkin's lymphoma (NHL) Crit Rev Oncol Hematol. 2001;40(1):37–51. doi: 10.1016/s1040-8428(01)00133-0. [DOI] [PubMed] [Google Scholar]
- 77.Knox S, Goris ML, Tempero M, Weiden PL, Gentner L, Breitz HB, Adams GP, Axworthy D, Gaffigan S, Bryan K, Fisher DR, Colcher D, Horak ID, Weiner LM. Phase II trial of 90Y-DOTA-biotin pre-targeted by NR-LU-10 antibody/streptavidin in patients with metastatic colon cancer. Clin Cancer Res. 2000;6:406–414. [PubMed] [Google Scholar]
- 78.Sharkey RM, Cardillo TM, Rossi EA, Chang CH, Karakay H, McBride WJ, Hansen HJ, Horak ID, Goldenberg DM. Signal amplification in molecular imaging by pre-targeting a multivalent, bispecific antibody. Nature Med. 2005;11:1250–1255. doi: 10.1038/nm1322. [DOI] [PubMed] [Google Scholar]
- 79.Roffler SR, Wang HE, Yu HM, Chang WD, Cheng CM, Lu YL, Chen BM, Cheng TL. A membrane antibody receptor for noninvasive imaging of gene expression. Gene Ther. 2006;13:412–420. doi: 10.1038/sj.gt.3302671. [DOI] [PubMed] [Google Scholar]
- 80.Behr TM, Sharkey RM, Juweid ME, Blumenthal RD, Dunn RM, Griffiths GL, Bair HJ, Wolf FG, Becker WS, Goldenberg DM. Reduction of the renal uptake of radiolabeled monoclonal antibody fragments by cationic amino acids and their derivatives. Cancer Res. 1995;55(17):3825–3834. [PubMed] [Google Scholar]
- 81.Adams GP, Schier R, McCall AM, Simmons HH, Horak EM, Alpaugh RK, Marks JD, Weiner LM. High affinity restricts the localization and tumor penetration of single-chain Fv antibody molecules. Cancer Res. 2001;61:4750–4755. [PubMed] [Google Scholar]
- 82.Jain M, Chauhan SC, Singh AP, Venkatraman G, Colcher D, Batra SK. Penetratin improves tumor retention of single-chain antibodies: a novel step towards optimization of radioimmunotherapy of solid tumors. Cancer Res. 2005;65(17):7840–7846. doi: 10.1158/0008-5472.CAN-05-0662. [DOI] [PubMed] [Google Scholar]
- 83.Vitetta ES, Ghetie VF. Considering therapeutic antibodies. Science. 2006;313:308–309. doi: 10.1126/science.1130482. [DOI] [PubMed] [Google Scholar]
- 84.Sharpe AH, Abbas AK. T-cell costimulation - biology, therapeutic potential, and challenges. N Engl J Med. 2006;355(10):973–975. doi: 10.1056/NEJMp068087. [DOI] [PubMed] [Google Scholar]
- 85.Suntharalingam G, Perry MR, Ward S, Brett SJ, Castello-Cortes A, Brunner MD, Panoskaltsis N. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med. 2006;355(10):1018–1028. doi: 10.1056/NEJMoa063842. [DOI] [PubMed] [Google Scholar]
- 86.Newsbriefs. NeutroSpec Withdrawn from Market. J Nucl Med. 2006;47(2):36N. [Google Scholar]
- 87.Albrecht H, DeNardo SJ. Recombinant antibodies: from the laboratory to the clinic. Cancer Biother Radiopharm. 2006;21(4):285–304. doi: 10.1089/cbr.2006.21.285. [DOI] [PubMed] [Google Scholar]
- 88.Reichert JM, Rosensweig CJ, Faden LB, Dewitz MC. Monoclonal antibody successes in the clinic. Nature Biotechnology. 2005;23(9):1073–1078. doi: 10.1038/nbt0905-1073. [DOI] [PubMed] [Google Scholar]
- 89.Reichert JM. Monoclonal antibodies in the clinic. Nature Biotechnol. 2001;19:819–822. doi: 10.1038/nbt0901-819. [DOI] [PubMed] [Google Scholar]
- 90.Reichert JM. Therapeutic monoclonal antibodies: trends in development and approval in the US. Curr Opin Mol Ther. 2002;4(2):110–118. [PubMed] [Google Scholar]
- 91.Holliger P, Hoogenboom H. Antibodies come back from the brink. Nature Biotechnol. 1998;16:1015–1016. doi: 10.1038/3460. [DOI] [PubMed] [Google Scholar]
- 92.Carter PJ. Potent antibody therapeutics by design. Nature Rev Immunol. 2006;6:343–357. doi: 10.1038/nri1837. [DOI] [PubMed] [Google Scholar]
- 93.Walsh G. Biopharmaceutical benchmarks 2006. Nature Biotechnol. 2006;24(7):769–776. doi: 10.1038/nbt0706-769. [DOI] [PubMed] [Google Scholar]
- 94.Wilkins DK, Mayer A. Development of antibodies for cancer therapy. Expert Opin Biol Ther. 2006;6(8):787–796. doi: 10.1517/14712598.6.8.787. [DOI] [PubMed] [Google Scholar]
- 95.Harris M. Monoclonal antibodies as therapeutic agents for cancer. The Lancet. 2004;5:292–302. doi: 10.1016/S1470-2045(04)01467-6. [DOI] [PubMed] [Google Scholar]
- 96.von Mehren M, Adams GP, Weiner LM. Monoclonal antibody therapy for cancer. Annu Rev Med. 2003;54:343–369. doi: 10.1146/annurev.med.54.101601.152442. [DOI] [PubMed] [Google Scholar]
- 97.Ross JS, Schenkein DP, Pietrusko R, Rolfe M, Linette GP, Stec J, Stagliano NE, Ginsburg GS, Symmans WF, Pusztai L, Hortobagyi GN. Targeted therapies for cancer 2004. Am J Clin Pathol. 2004;122:598–609. doi: 10.1309/5CWP-U41A-FR1V-YM3F. [DOI] [PubMed] [Google Scholar]
- 98.White CA, Weaver RL, Grillo-Lopez AJ. Antibody-targeted immunotherapy for treatment of malignancy. Annu Rev Med. 2001;52:125–145. doi: 10.1146/annurev.med.52.1.125. [DOI] [PubMed] [Google Scholar]
- 99.Oriuchi H, Higuchi T, Hanaoka H, Iida Y, Endo K. Current status of cancer therapy with radiolabeled monoclonal antibody. Ann Nucl Med. 2005;19(5):355–365. doi: 10.1007/BF03027399. [DOI] [PubMed] [Google Scholar]
- 100.Ross JS, Gray K, Gray GS, Worland PJRM. Anticancer antibodies. Am J Clin Pathol. 2003;119(4):472–485. doi: 10.1309/Y6LP-C0LR-726L-9DX9. [DOI] [PubMed] [Google Scholar]
- 101.Reichert JM, Pavlou A. Monoclonal antibodies market. Nature Rev Drug Disc. 2004;3:383–384. doi: 10.1038/nrd1386. [DOI] [PubMed] [Google Scholar]
- 102.Pavlou AK, Reichert JM. Recombinant protein therapeutics -- success rates, market trends, and values to 2010. Nat Biotech. 2004;22:1513. doi: 10.1038/nbt1204-1513. [DOI] [PubMed] [Google Scholar]
- 103.Saijo N. What are the reasons for negative phase III trials of molecular-target-based drugs? Cancer Sci. 2004;95(10):772–776. doi: 10.1111/j.1349-7006.2004.tb02180.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Wu AM, Senter PD. Arming antibodies: prospects and challenges for immunoconjugates. Nature Biotechnol. 2005;23(9):1137–1146. doi: 10.1038/nbt1141. [DOI] [PubMed] [Google Scholar]
- 105.Sharkey RM, Goldenberg DM. Targeted therapy of cancer: new prospects for antibodies and immunoconjugates. CA Cancer J Clin. 2006;56:226–243. doi: 10.3322/canjclin.56.4.226. [DOI] [PubMed] [Google Scholar]
- 106.Spicer J. Technology evaluation: Nimotuzumab, the Center of Molecular Immunology/YM BioSciences/Oncoscience. Curr Opin Mol Ther. 2005;7(2):182–191. [PubMed] [Google Scholar]
- 107.Casaco A. Loco-regional radioimmunotherapy of high grade malignant gliomas using the humanized monoclonal antibody, h-R3, labeled with 188Re. Proc Am Soc Clin Oncol. 2004;22 Abs 2530. [Google Scholar]
- 108.Vallis KA, Reilly RM, Chen P, Oza A, Hendler A, Cameron R, Hershkop M, Iznaga-Escobar N, Ramos-Suzarte M, Keane P. 99mTc-hR3 (DiaCIM), a humanized immunoconjugate directed towards the epidermal growth factor receptor. Nucl Med Commun. 2002;23(12):1155–1164. doi: 10.1097/00006231-200212000-00002. [DOI] [PubMed] [Google Scholar]
- 109.Divgi C. What ails solid tumor radioimmunotherapy? Cancer Biother Radiopharm. 2006;21(2):81–84. doi: 10.1089/cbr.2006.21.81. [DOI] [PubMed] [Google Scholar]
- 110.Blumenthal RD, Kashi R, Sharkey RM, Goldenberg DM. Quantitative and qualitative effects of experimental radioimmunotherapy on tumor vascular permeability. Int J Cancer. 1995;61(4):557–566. doi: 10.1002/ijc.2910610421. [DOI] [PubMed] [Google Scholar]
- 111.DeNardo GJ, Kennel SJ, Siegel JA, DeNardo SJ. Radiometals as payloads for radioimmunotherapy for lymphoma. Clin Lymphoma. 2004;5(S1):S5–S10. doi: 10.3816/clm.2004.s.002. [DOI] [PubMed] [Google Scholar]
- 112.Vose JM. Bexxar: Novel radioimmunotherapy for the treatment of low-grade and transformed low-grade non-Hodgkin's lymphoma. The Oncologist. 2004;9:160–172. doi: 10.1634/theoncologist.9-2-160. [DOI] [PubMed] [Google Scholar]
- 113.Linden O, Hindorf C, Cavallin-Stahl E, Wegener WA, Goldenberg DM, Horne H, Ohlsson T, Stenberg L, Strand SE, Tennvall J. Dose-fractionated radioimmunotherapy in non-Hodgkin's lymphoma using DOTA-conjugated, 90Y-radiolabeled, humanized anti-CD22 monoclonal antibody, epratuzumab. Clin Cancer Res. 2005;11(14):5215–5222. doi: 10.1158/1078-0432.CCR-05-0172. [DOI] [PubMed] [Google Scholar]
- 114.Immunomedics Inc. Treatment of NHL with 90Y-hLL2 IgG. Accessed February 2007 at http://www.clinicaltrials.gov/ct/show/NCT00421395.
- 115.Lamonica D, Czuczman M, Nabi H, Klippenstein D, Grossman Z. Radioimmunoscintigraphy (RIS) with bectumomab (99mTc-Labeled IMMU-LL2, Lymphoscan) in the assessment of recurrent non-Hodgkin's lymphoma (NHL) Cancer Biother Radiopharm. 2002;17(6):689–697. doi: 10.1089/108497802320970307. [DOI] [PubMed] [Google Scholar]
- 116.Blend MJ, Hyun H, Kozloff M, Levi H, Mills GQ, Gasparini M, Buraggi G, Hughes L, Pinsky CM, Goldenberg DM. Improved staging of B-cell non-Hodgkins lymphoma patients with 99mTc-labeled LL2 monoclonal antibody fragment. Cancer Res. 1995;55(23 Suppl S):S5764–S5770. [PubMed] [Google Scholar]
- 117.DeNardo GJ, Kukis DL, Shen S, DeNardo DJ, Meares CF, DeNardo SJ. 67Cu- versus 131I-labeled Lym-1 antibody: comparative pharmacokinetics and dosimetry in patients with non-Hodgkin's lymphoma. Clin Cancer Res. 1999;5:533–541. [PubMed] [Google Scholar]
- 118.Peregrine Pharmaceuticals Inc. Prospectus - September 29, 2005. Accessed Feb. 2007; follow hyperlink to peregrine42b2.htm at http://www.sec.gov/Archives/edgar/data/704562/000101968705002707/
- 119.Belch A, McEwan A, Hewitt J, Riauka T, Stabin M, Brill A, Gallant G, Blake L, Klein J. Tumor targeting, dosimetry and clinical response data for lymphorad-131 in patients with relapsed/refractory non-Hodgkin's lymphoma. Blood. 2004;104(11):214A–215A. [Google Scholar]
- 120.Beatty JD, Williams LE, Yamauchi D, Morton BA, Hill LR, Beatty BG, Paxton RJ, Merchant B, Shively JE. Presurgical imaging with indium-labeled anti-carcinoembryonic antigen for colon cancer staging. Cancer Res. 1990;50:922s–926s. [PubMed] [Google Scholar]
- 121.Bischof-Delaloye A, Delaloye B, Buchegger F, Gilgien W, Studer A, Curchod S, Givel JC, Mosimann F, Pettavel J, Mach JP. Clinical value of immunoscintigraphy in colorectal carcinoma patients: a prospective study. J Nucl Med. 1989;30:1646–1656. [PubMed] [Google Scholar]
- 122.Blumenthal RD, Sharkey RM, Kashi R, Goldenberg DM. Comparison of therapeutic efficacy and host toxicity of two different 131I-labeled antibodies and their fragments in the GW-39 colonic cancer xenograft model. Int J Cancer. 1989;44:292–300. doi: 10.1002/ijc.2910440218. [DOI] [PubMed] [Google Scholar]
- 123.Scott AM, Lee FT, Jones R, Hopkins W, MacGregor D, Cebon JS, Hannah A, Chong G, U P, Papenfuss A, Rigopoulos A, Sturrock S, Murphy R, Wirth V, Murone C, Smyth FE, Knight S, Welt S, Ritter G, Richards E, Nice EC, Burgess AW, Old LJ. A phase I trial of humanized monoclonal antibody A33 in patients with colorectal carcinoma: biodistribution, pharmacokinetics, and quantitative tumor uptake. Clin Cancer Res. 2005;11:4810–4817. doi: 10.1158/1078-0432.CCR-04-2329. [DOI] [PubMed] [Google Scholar]
- 124.Chong G, Lee FT, Hopkins W, Tebbut N, Cebon JS, Mountain AJ, Chappell B, Papenfuss A, Schleyer P, U P, Murphy R, Wirth V, Smyth FE, Potasz N, Poon A, Davis ID, Saunder T, O'Keefe GJ, Burgess AW, Hoffman EW, Old LJ, Scott AM. Phase I trial of 131I-huA33 in patients with advanced colorectal carcinoma. Clin Cancer Res. 2005;11:4818–4826. doi: 10.1158/1078-0432.CCR-04-2330. [DOI] [PubMed] [Google Scholar]
- 125.Buchsbaum D, Khazaeli MB, Liu TP, Bright S, Richardson K, Jones M, Meredith R. Fractionated radioimmunotherapy of human colon carcinoma xenografts with 131I-labeled monoclonal antibody CC49. Cancer Res. 1995;55(23 Suppl S):S5881–S5887. [PubMed] [Google Scholar]
- 126.Tempero M, Leichner P, Baranowska-Kortylewicz J, Harrison K, Augustine S, Schlom J, Anderson J, Wisecarver J, Colcher D. High-dose therapy with 90Yttrium-labeled monoclonal antibody CC49: a phase I trial. Clin Cancer Res. 2000;6:3095–3102. [PubMed] [Google Scholar]
- 127.Gansow OA, Wu C, Goldenberg DM. Cancer therapy with radiolabeled antibodies. CRC Press; 1995. Advanced methods for radiolabeling monoclonal antibodies with therapeutic radionuclides; pp. 63–76. [Google Scholar]
- 128.Immunomedics Inc. 10-K for IMMUNOMEDICS, INC - Aug. 29, 2006. Accessed Feb. 2007 at http://www.sec.gov/Archives/edgar/data/722830/000119312506181414/d10k.htm.
- 129.Immunomedics Inc. Safety study of hMN14 to treat either colorectal or breast cancer. Accessed Feb. 2007 at www.clinicaltrials.gov/ct/show/NCT00041652.
- 130.Immunomedics Inc. Safety Study of 90Y-hMN14 to Treat Pancreatic Cancer. Accessed Feb. 2007 at http://www.clinicaltrials.gov/ct/show/NCT00041639.
- 131.Liersch T, Meller J, Kulle B, Behr TM, Markus P, Langer C, Ghadimi BM, Wegener WA, Kovacs J, Horak ID, Becker H, Goldenberg DM. Phase II trial of carcinoembryonic antigen radioimmunotherapy with 131I-labetuzumab after salvage resection of colorectal metastases in the liver: five-year safety and efficacy results. J Clin Oncol. 2005;23(27):6763–6770. doi: 10.1200/JCO.2005.18.622. [DOI] [PubMed] [Google Scholar]
- 132.Immunomedics Inc. Safety Study of 90Y-hMN14 to treat colorectal cancer patients with limited residual disease after surgery. Accessed Feb. 2007 at http://www.clinicaltrials.gov/ct/show/NCT00041691.
- 133.Wong JYC, Shibata S, Williams LE, Kwok CS, Liu A, Chu DZ, Yamauchi DM, Wilczynski S, Ikle DN, Wu AM, Yazaki PJ, Shively JE, Doroshow JH, Raubitschek AA. A phase I trial of 90Y-anticarcinoembryonic antigen chimeric T84.66 radioimmunotherapy with 5-fluorouracil in patients with metastatic colorectal cancer. Clin Cancer Res. 2003;9:5842–5852. [PubMed] [Google Scholar]
- 134.DeNardo SJ, L KE, O'Donnell RT, Richman CM, Salako QA, Shen S, Noz M, Glenn SD, Ceriani RL, deNardo GJ. Radioimmunotherapy for breast cancer using 111In/90Y-BrE-3: results of a phase I clinical trial. J Nucl Med. 1997;38(8):1180–1185. [PubMed] [Google Scholar]
- 135.DeNardo SJ, Richman CM, Goldstein DS, et al. Yttrium-90/Indium-111- DOTA-peptide-chimeric L6: pharmacokinetics, dosimetry and initial results in patients with incurable breast cancer. Anticancer Res. 1997;17:1735–1744. [PubMed] [Google Scholar]
- 136.Richman CM, DeNardo SJ, O'Donnell RT, Yuan A, Natarajan A, Wun T, Tuscano JM, Chew HL, Lara PN, deNardo GJ. Combined modality radioimmunotherapy (RIT) in metastatic prostate and breast cancer using paclitaxel and a MUC-1 monoclonal antibody, m170, linked to 90Y: a phase I trial. J Clin Oncol. 2004;22(14S):2554. [Google Scholar]
- 137.Schrier DM, Stemmer SM, Johnson T, Kasliwal R, Lear J, Matthes S, Taffs S, Dufton C, Glenn SD, Butchko G, Ceriani RL, Rovira D, Bunn P, Shpall EJ, Bearman SI, Purdy M, Cagnoni M, Jones RB. High-dose 90Y-Mx-diethylenetriaminepentaacetic acid (DTPA)-BrE-3 and autologous hematopoietic stem cell support (AHSCS) for the treatment of advanced breast cancer: A phase I trial. Cancer Res. 1995;55:5921s–5924s. [PubMed] [Google Scholar]
- 138.Ebeling FG, Stieber P, Untch M, Nagel D, Konecny GE, Schmitt UM, Fateh-Moghadam A, Seidel D. Serum CEA and CA 15-3 as prognostic factors in primary breast cancer. Br J Cancer. 2002;86(8):1217–1222. doi: 10.1038/sj.bjc.6600248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Behr TM, Sharkey RM, Juweid ME, Dunn RM, Vagg RC, Ying Z, Zhang CH, Swayne LC, Vardi Y, Siegel JA, Goldenberg DM. Phase I/II clinical radioimmunotherapy with an 131I-labeled anti-carcinoembryonic antigen murine monoclonal antibody IgG. J Nucl Med. 1997;38(6):858–870. [PubMed] [Google Scholar]
- 140.Sanidas EE, Koukouraki S, Velidaki A, Manios A, Stathopoulos E, De Bree I, Kafousi M, Kodogiannis E, Karkavitsas N, Tsiftsis DD. Contribution of 99mTc-anti-carinoembryonic antigen antibody and 99mTc-sestamibi scintimammograpy in the evaluation of high risk palpable breast lesions. Nucl Med Commun. 2003;24(3):291–296. doi: 10.1097/00006231-200303000-00009. [DOI] [PubMed] [Google Scholar]
- 141.DeNardo SJ, Kukis DL, Miers LA, Winthrop MD, Kroger LA, Salako QA. Yttrium-90-DOTA-peptide ChL6 radioimmunoconjugate: efficacy and toxicity in mice bearing p53 mutant human breast cancer xenografts. J Nucl Med. 1998;39:842–849. [PubMed] [Google Scholar]
- 142.Burke PA, DeNardo SJ, Miers L, Lamborn KR, Matzku S, DeNardo GL. Cilengitide targeting of alpha-v-beta-3 integrin receptor synergizes with radioimmunotherapy to increase efficacy and apoptosis in breast cancer xenografts. Cancer Res. 2002;62:4263–4272. [PubMed] [Google Scholar]
- 143.DeNardo SJ, Kukis DL, Kroger LA, O'Donnell RT, Lamborn KR, Miers LA, deNardo DJ, Meares CF, deNardo GJ. Synergy of taxol and radioimmunoimmunotherapy with 90Y-labeled chimeric L6 antibody: efficacy and toxicity in breast cancer xenografts. Proc Natl Acad Sci USA. 1997;94:4000–4004. doi: 10.1073/pnas.94.8.4000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Burke PA, deNardo SJ, Miers LA, Kukis DL, deNardo GJ. Combined modality radioimmunotherapy: promise and peril. Cancer. 2002;94(S4):1320–1331. doi: 10.1002/cncr.10303. [DOI] [PubMed] [Google Scholar]
- 145.Bander NH. Technology insight: monoclonal antibody imaging of prostate cancer. Nature Clin Pract. 2006;3(4):216–225. doi: 10.1038/ncpuro0452. [DOI] [PubMed] [Google Scholar]
- 146.Raj GV, Partin AW, Polascik TJ. Clinical utility of 111In-capromab pendetide immunoscintigraphy in the detection of early, recurrent prostate carcinoma after radical prostatectomy. Cancer. 2002;94(4):987–996. [PubMed] [Google Scholar]
- 147.Meredith RF, Bueschen AJ, Khazaeli MB, Plott WE, Grizzle WE, Wheeler RH, Schlom J, Russel CD, Liu T, LoBuglio AF. Treatment of metastatic prostate carcinoma with radiolabeled antibody CC49. J Nucl Med. 1994;35(6):1017–1022. [PubMed] [Google Scholar]
- 148.Vallabhajosula S, Goldsmith SJ, Kostakoglu L, Milowsky MI, Nanus DM, Bander NH. Radioimmunotherapy of prostate cancer using 90Y- and 177Lu-labeled J591 monoclonal antibodies: effect of multiple treatments on myelotoxicity. Clin Cancer Res. 2005;11(19 Pt 2):7195s–7200s. doi: 10.1158/1078-0432.CCR-1004-0023. [DOI] [PubMed] [Google Scholar]
- 149.O'Donnell RT, deNardo SJ, Miers LA, Lamborn KR, Kukis DL, deNardo GJ, Meyers FJ. Combined modality radioimmunotherapy for human prostate cancer xenografts with taxanes and 90Y-DOTA-peptide-ChL6. The Prostate. 2002;50(1):27–37. doi: 10.1002/pros.10029. [DOI] [PubMed] [Google Scholar]
- 150.Nicodemus CF, Berek JS. Monoclonal antibody therapy of ovarian cancer. Expert Rev Anticancer Ther. 2005;5(1):87–96. doi: 10.1586/14737140.5.1.87. [DOI] [PubMed] [Google Scholar]
- 151.Epenetos AA, Hird V, Lambert H, Mason P, Coulter C. Long term survival of patients with advanced ovarian cancer treated with intraperitoneal radioimmunotherapy. Int J Gynecoll Cancer. 2000;10 Suppl. 1:44–46. doi: 10.1046/j.1525-1438.2000.99510.x. [DOI] [PubMed] [Google Scholar]
- 152.Seiden MV, Benigno BB. Group, T. S. o. M. A. R. S., A pivotal phase III trial to evaluate the efficacy and safety of adjuvant treatment with 90Y-labeled monoclonal antibody, HMFGI, in ovarian cancer. Proc Am Soc Clin Oncol. 2004;23 (suppl):16a. Abstract 5008. [Google Scholar]
- 153.Buist MR, Molthoff CFM, Kenemans P, Meijer CJLM. Distribution of Ov-Tl 3 and Mov18 in normal and malignant ovarian tissue. J Clin Pathol. 1995;48(7):631–636. doi: 10.1136/jcp.48.7.631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Buijs WC, Tibben JG, Boerman OC, Molthoff CF, Massuger LF, Koenders EB, Schijf CP, Siegel JA, Corstens FH. Dosimetric analysis of chimeric monoclonal antibody cMOv18 IgG in ovarian carcinoma patients after intraperitoneal and intravenous administration. Eur J Nucl Med. 1998;25(11):1552–1561. doi: 10.1007/s002590050335. [DOI] [PubMed] [Google Scholar]
- 155.Coliva A, Zacchetti A, Luison E, Tomassetti A, Bongarzone I, Seregni E, Bombardieri E, Martin F, Giussani A, Figini M, Canevari S. 90Y labeling of monoclonal antibody MOv18 and pre-clinical validation for radioimmunotherapy of human ovarian carcinomas. Cancer Immunol Immunother. 2005;54:1200–1213. doi: 10.1007/s00262-005-0693-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Breitz HB, Tyler A, Bjorn MJ, Lesley T, Weiden PL. Clinical experience with 99mTc-nofetumomab merpentan (Verluma) radioimmunoscintigraphy. Clin Nucl Ned. 1997;22(9):615–620. doi: 10.1097/00003072-199709000-00007. [DOI] [PubMed] [Google Scholar]
- 157.Machac J, Krynyckyi B, Kim C. Peptide and antibody imaging in lung cancer. Semin Nucl Med. 2002;32(4):276–292. doi: 10.1053/snuc.2002.126053. [DOI] [PubMed] [Google Scholar]
- 158.Forero A, Meredith RF, Khazaeli MB, Shen S, Grizzle WE, Carey D, Busby E, LoBuglio AF, Robert F. Phase I study of 90Y-CC49 monoclonal antibody therapy in patients with advanced non-small cell lung cancer: effect of chelating agents and paclitaxel administration. Cancer Biother Radiopharm. 2005;20(5):467–478. doi: 10.1089/cbr.2005.20.467. [DOI] [PubMed] [Google Scholar]
- 159.Arista A, Sturiale C, Riva P, Tison V, Frattarelli M, Moscatelli G, Franceschi G, Spinelli A. Intralesional administration of 131I-labeled monoclonal antibodies in the treatment of malignant gliomas. Acta Neurochirurgica. 1995;135(3–4):159–162. doi: 10.1007/BF02187762. [DOI] [PubMed] [Google Scholar]
- 160.Shapiro WR, Carpenter SP, Roberts K, Shan JS. 131I-chTNT-1/B mAb: tumor necrosis therapy for malignant astrocytic glioma. Expert Opin Biol Ther. 2006;6(5):539–545. doi: 10.1517/14712598.6.5.539. [DOI] [PubMed] [Google Scholar]
- 161.Bonino LD, Demonte LB, Spagnoli GC, Vola R, Mariani M, Barone D, Moro AM, Riva P, Nicotra MR, Natali PG, Malavasi F. Bispecific monoclonal antibody anti-Cd3 × anti-tenascin - an immunotherapeutic agent for human glioma. Int J Cancer. 1995;61(4):509–515. doi: 10.1002/ijc.2910610414. [DOI] [PubMed] [Google Scholar]
- 162.Bigner DD, Brown M, Coleman RE, Friedman AH, Friedman HS, McLendon RE, Bigner SH, Zhao XG, Wikstrand CJ, Pegram CN, Kerby T, Zalutsky MR. Phase I studies of treatment of malignant gliomas and neoplastic meningitis with 131I-radiolabeled monoclonal antibodies anti-tenascin 81C6 and anti-chondroitin proteoglycan Mel-14 F(ab')2 preliminary report. J.Neuro-Oncol. 1995;24:109–122. doi: 10.1007/BF01052668. [DOI] [PubMed] [Google Scholar]
- 163.Reardon D, Akabani G, Coleman RE, Friedman AH, Herndon J, Cokgor I, McLendon R, Pegram CN, Provenzale JM, Quinn JA, Rich JN, Regalado LV, Sampson JH, Shafman DT, Wikstrand CJ, Wong TZ, Zhao XG, Zalutsky MR, Bigner D. Phase II trial of murine 131I-labeled antitenascin monoclonal antibody 81C6 administered into surgically created resection cavities of patients with newly diagnosed malignant gliomas. J Clin Oncol. 2002;20(5 March 1):1389–1397. doi: 10.1200/JCO.2002.20.5.1389. [DOI] [PubMed] [Google Scholar]
- 164.Akabani G, Reardon DA, Coleman RE, Wong TZ, Metzler SD, Bowsher JE, Barboriak DP, Provenzale JM, Greer KL, DeLong D. Dosimetry and radiographic analysis of 131I-labeled anti-tenascin 81C6 murine monoclonal antibody in newly diagnosed patients with malignant gliomas: a phase II study. J Nucl Med. 2005;46(6):1042–1051. [PubMed] [Google Scholar]
- 165.Paganelli G, Bartolomei M, Ferrari M, Cremonesi M, Broggi G, Maira G, Sturiale C, Grana C, Prisco G, Gatti M, Caliceti P, Chinol M. Pre-targeted locoregional radioimmunotherapy with 90Y-biotin in glioma patients: Phase I study and preliminary therapeutic results. Cancer Biother Radiopharm. 2001;16(3):227–235. doi: 10.1089/10849780152389410. [DOI] [PubMed] [Google Scholar]
- 166.Grana C, Chinol M, Robertson C, Mazzetta C, Bartolomei M, de Cicco C, Fiorenza M, Gatti M, Caliceti P, Paganelli G. Pre-targeted adjuvant radioimmunotherapy with 90Y-biotin in malignant glioma patients: A pilot study. Br J Cancer. 2002;86:207–212. doi: 10.1038/sj.bjc.6600047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.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 131I-labeled monoclonal antibody G250 in metastatic renal cell carcinoma. Clin Cancer Res. 1998;4:2729–2739. [PubMed] [Google Scholar]
- 168.Divgi CR, O'Donoghue JA, Welt S, O'Neel J, Finn RD, 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(8):1412–1421. [PubMed] [Google Scholar]
- 169.Goldenberg DM. Advancing role of radiolabeled antibodies in the therapy of cancer. Cancer Immunol Immunother. 2003;52:281–296. doi: 10.1007/s00262-002-0348-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Projection based on Sg2 Imaging Forcast, U. M.-S. Skokie, Il: www.sg2.com) [Google Scholar]
- 171.Kang DE, White RL, Jr, Zuger JH, Sasser HC, Teigland CM. Clinical use of fluorodeoxyglucose 18F positron emission tomography for detection of renal cell carcinoma. J Urol. 2004;171:1806–1809. doi: 10.1097/01.ju.0000120241.50061.e4. [DOI] [PubMed] [Google Scholar]
- 172.Libutti SK, Alexander HR, Jr, Choyke PL, Bartlett DL, Bacharach SL, Whatley M, Jousse F, Eckelman WC, Kranda K, Neumann RD, Carrasquillo JA. A prospective study of 2-[18F] fluoro-2-deoxy-Dglucose/ positron emission tomography scan, 99mTc-labeled arcitumomab (CEAScan), and blind second-look laparotomy for detecting colon cancer recurrence in patients with increasing carcinoembryonic antigen levels. Ann Surg Oncol. 2001;8:779–786. doi: 10.1007/s10434-001-0779-9. [DOI] [PubMed] [Google Scholar]
- 173.Wittes RE. Cancer weapons, out of reach. The Washington Post; 2004. Jun 28, [Google Scholar]
- 174.Choyke PL. Invited commentary. RadioGraphics. 2006;26:1806–1810. [Google Scholar]
- 175.Kocher DC. Radioactive decay tables. Office of Scientific and Technical Information, U.S. Department of Energy; 1981. [Google Scholar]
- 176.Katz L, Penfold AS. Range-energy relations for electrons and the determination of beta-ray end-point energies by absorption. Rev Mod Phys. 1952;24:28–44. [Google Scholar]