MedPath

212Pb-DOTAM-GRPR1 Advanced Drug Monograph

Published:Jun 9, 2025

Generic Name

212Pb-DOTAM-GRPR1

Comprehensive Report on the Investigational Radiopharmaceutical 212Pb-DOTAM-GRPR1

I. Introduction to 212Pb-DOTAM-GRPR1: A Novel Targeted Alpha Therapy

A. Overview of the Radiopharmaceutical

212Pb-DOTAM-GRPR1, also designated Pb-GRPR, is an investigational radioimmunoconjugate currently under clinical evaluation for cancer treatment.[1] This therapeutic agent is meticulously designed for targeted alpha therapy (TAT), a sophisticated strategy aiming to deliver potent cytotoxic radiation directly to malignant cells. The fundamental structure of 212Pb-DOTAM-GRPR1 comprises three key components: the alpha-emitting radionuclide lead-212 ($^{212}$Pb), the macrocyclic metal chelator DOTAM (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic amide), and a peptide antagonist that specifically targets the Gastrin-Releasing Peptide Receptor (GRPR).[1] The nomenclature of this compound, with the alternative designation "Pb-GRPR," appears to reflect an effort to simplify its complex chemical name for clearer communication within clinical and broader scientific forums. This radioconjugate is engineered with the intent of exerting antineoplastic activity by selectively binding to and irradiating tumor cells that overexpress GRPR.[2] The core principle of TAT, as embodied by 212Pb-DOTAM-GRPR1, involves the precise conjugation of a biological targeting molecule with a highly energetic, short-range alpha-emitting radioisotope, thereby concentrating the therapeutic effect within the tumor microenvironment.[4]

B. Significance in the Landscape of Cancer Treatment

Targeted Alpha Therapies, including 212Pb-DOTAM-GRPR1, represent an innovative and promising frontier in oncological research, particularly for malignancies that have proven refractory to conventional treatments or have developed resistance mechanisms.[2] The defining characteristic of alpha particles, their high linear energy transfer (LET) and remarkably short path length in biological tissues, underpins the therapeutic rationale for TAT. This combination offers the potential for exceptionally potent tumor cell eradication while concurrently minimizing radiotoxic damage to adjacent healthy tissues—a critical advantage over therapies employing beta-emitting radionuclides or traditional systemic chemotherapy agents.[2] The high LET of alpha particles, typically around 100 keV/µm, translates into dense ionization tracks that cause complex, irreparable DNA damage, such as double-strand breaks, within the targeted cells.[4] The short range, on the order of micrometers, confines this intense cytotoxic effect primarily to the tumor cells and their immediate microenvironment, thereby enhancing the therapeutic index. This localized and potent cell-killing mechanism is central to the promise of TAT.

The strategic selection of GRPR as the molecular target further amplifies the potential clinical utility of 212Pb-DOTAM-GRPR1, given that GRPR is documented to be overexpressed in a diverse array of solid tumors.[1] This broad expression profile suggests a wide spectrum of cancers that could potentially be amenable to this targeted approach. The development of 212Pb-DOTAM-GRPR1 is notably driven by the pressing need to address unmet medical needs, particularly for patients with advanced or refractory cancers where existing therapeutic options are exhausted or inadequate.[2] The unique mechanism of action of alpha emitters, involving direct and severe DNA damage, holds the potential to overcome certain acquired resistance mechanisms observed with other therapies.[4]

II. Molecular Profile and Components of 212Pb-DOTAM-GRPR1

The efficacy and safety of 212Pb-DOTAM-GRPR1 are intrinsically linked to the distinct properties of its constituent parts: the $^{212}$Pb radioisotope, the DOTAM chelator, and the GRPR1 antagonist peptide.

### A. The Alpha-Emitting Radioisotope: Lead-212 ($^{212}Pb)∗∗RadiophysicalPropertiesandDecayCascade:∗∗Lead−212(^{212}$Pb) is a radioisotope with a physical half-life of approximately 10.6 hours.2 This intermediate half-life is considered advantageous for targeted radionuclide therapy, providing a sufficient window for systemic administration, biodistribution to tumor sites, and therapeutic action before substantial radioactive decay and biological clearance occur.2 While $^{212}Pbitselfundergoesbeta(\beta^-)decay,itservesasan∗invivo∗generatorofhighlypotentalpha(\alpha$) particles. Its decay product, Bismuth-212 ($^{212}Bi),withahalf−lifeofapproximately60.6minutes,subsequentlydecaysviaalphaemission(approximately36^{212}$Po), which has an extremely short half-life (~0.3 µs) and decays almost exclusively by alpha emission.10 It is these sequential alpha emissions from $^{212}$Bi and $^{212}$Po that are responsible for the primary cytotoxic effect of $^{212}$Pb-based TAT. Alpha particles are characterized by a high LET, typically around 100 keV/µm, and a very short range in tissue, generally 50-100 µm, equivalent to only a few cell diameters.4 The "in vivo generator" characteristic of $^{212}$Pb, where the therapeutically active alpha-emitting daughters are produced at the tumor site following the localization of the $^{212}$Pb-radiopharmaceutical, is a key feature. However, this also introduces complexities concerning the fate of these daughter nuclides. The recoil energy from alpha decay and the change in elemental identity could potentially destabilize the chelation complex. If daughter radionuclides like $^{212}$Bi or $^{212}$Po are released from the chelator post-decay, they could migrate and cause unintended off-target radiotoxicity. This underscores the paramount importance of employing an exceptionally stable chelator capable of retaining both the parent $^{212}$Pb and its decay progeny.

Rationale for Use in Targeted Alpha Therapy:

The rationale for utilizing $^{212}$Pb in TAT is compelling. The high LET of the emitted alpha particles results in dense ionization clusters along their path through tissue. This concentrated energy deposition leads to complex and highly lethal DNA damage, particularly irreparable double-strand breaks (DSBs), within the nucleus of targeted cells.4 The cytotoxicity is so profound that it is estimated only a few alpha particle traversals through a cell nucleus are sufficient to induce cell death.4 Furthermore, the short range of these alpha particles is a critical attribute, ensuring that their destructive energy is predominantly confined to the targeted tumor cells and the immediate tumor microenvironment, thereby minimizing radiation exposure to distant, healthy tissues and organs.4 This localized energy deposition makes $^{212}$Pb particularly well-suited for eradicating micrometastases, which are often inaccessible or resistant to other therapeutic modalities.4 An additional advantage is that the direct and severe DNA damage inflicted by alpha particles may circumvent common mechanisms of resistance to other cancer therapies and, importantly, does not necessarily require internalization of the targeting vector into the cell to be effective, as the alpha particles can traverse cellular compartments to reach the nucleus.4

Production and Availability:

$^{212}Pbisamemberofthethoriumdecayseriesandcanbeobtainedfromradionuclidegeneratorsystemscontaininglonger−livedparentisotopes,suchasThorium−228(^{228}Th,half−life1.9years)orRadium−224(^{224}$Ra, half-life 3.6 days).10 Orano Med, the developer of 212Pb-DOTAM-GRPR1, has made significant investments in establishing robust capabilities for the production of high-purity $^{212}$Pb. These include specialized industrial-scale facilities designed to ensure a consistent and reliable supply of the radioisotope for ongoing clinical trials and anticipated future commercial demands.2 The relatively short half-life of $^{212}$Pb necessitates efficient production processes and well-coordinated logistics, often requiring radiolabeling procedures to be performed on-site at or near the clinical facility where the drug will be administered.10

B. The Chelating Agent: DOTAM (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic amide; TCMC)

Chemical Structure and Function:

DOTAM, also known by the chemical name 1,4,7,10-tetrakis(carbamoylmethyl)-1,4,7,10-tetraazacyclododecane and sometimes referred to as TCMC, is a macrocyclic chelating agent.3 Its primary role within the 212Pb-DOTAM-GRPR1 construct is to form a highly stable complex with the $^{212}$Pb radioisotope. This chelation is crucial for securely attaching the radioactive payload to the GRPR1 targeting peptide and preventing its premature dissociation and non-specific distribution within the body, which could lead to off-target toxicity.3

Advantages for $^{212}$Pb Chelation and Stability:

The selection of DOTAM for $^{212}$Pb-based radiopharmaceuticals is a deliberate and significant choice. Comparative studies have indicated that DOTAM exhibits superior chelation efficiency and faster radiolabeling kinetics with $^{212}$Pb compared to other chelators, including the widely used DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid).12 While DOTA is an excellent chelator for various radiometals like $^{177}$Lu and $^{68}$Ga, it has been found to be suboptimal for $^{212}$Pb.12 The development and adoption of DOTAM specifically addressed this deficiency, providing a more robust platform for $^{212}$Pb. DOTAM forms a particularly stable complex with $^{212}$Pb, a property that is essential for maintaining the integrity of the radiopharmaceutical in vivo, ensuring effective delivery of the radioisotope to the tumor target, and minimizing systemic exposure to free $^{212}$Pb.4 The overall charge of the metal-chelate complex can also influence its biodistribution; the $^{212}$Pb-DOTAM complex carries a +2 charge, which may affect its interaction with biological tissues, including the kidneys.12 This careful selection and optimization of the chelator underscores a nuanced understanding of radiometal chemistry tailored to the specific properties of $^{212}$Pb, aiming to maximize both the stability and biological performance of the resulting radiopharmaceutical.

C. The Targeting Ligand: GRPR1 Antagonist

Structure and Specificity for GRPR:

The targeting component of 212Pb-DOTAM-GRPR1 is a peptide antagonist engineered to bind with high specificity and affinity to the Gastrin-Releasing Peptide Receptor (GRPR).1 While the precise amino acid sequence of the proprietary peptide designated "GRPR1" used in this specific radiopharmaceutical is not disclosed in the available documentation 7, it is designed to recognize and dock with GRPR expressed on the surface of cancer cells. The lack of specific structural information for the GRPR1 peptide is a limitation for a complete molecular characterization based solely on the provided materials, as the peptide's sequence dictates its binding characteristics and pharmacokinetic behavior. General structures of other GRPR antagonists have been reported in the literature 17, but these are not explicitly linked to the GRPR1 component of 212Pb-DOTAM-GRPR1.

Rationale for Antagonist vs. Agonist Approach:

The choice of a GRPR antagonist over an agonist for this radioligand therapy is a strategic decision rooted in pharmacological considerations. GRPR antagonists have demonstrated several advantages in the context of targeted radionuclide delivery. Preclinical and clinical observations suggest that antagonists can achieve enhanced tumor accumulation and prolonged retention times within the tumor tissue compared to their agonist counterparts.2 Furthermore, antagonists often exhibit lower uptake and faster clearance from normal physiological tissues that also express GRPR (such as the pancreas), which can lead to a more favorable therapeutic index.2 One proposed reason for this improved performance is that the binding of antagonists may be independent of the receptor's coupling state with G-proteins, potentially allowing them to bind to a larger pool of available receptor sites on the tumor cell surface.12 This implies that GRPR antagonists might offer a more sustained and selective tumor-targeting profile, which is highly advantageous when delivering a potent radiotoxic payload like $^{212}$Pb. This preference for antagonists reflects an optimization strategy aimed at maximizing tumor radiation dose while minimizing systemic toxicity.

### Table 1: Key Characteristics of 212Pb-DOTAM-GRPR1 Components

| Component | Name | Half-life | Emissions | Alpha Particle Energy/Range | Key Features |

|-------------------|-------------------------------------------------------------------------------|--------------|---------------------------------------------------------------------------|--------------------------------------|----------------------------------------------------------------------------------------------------------|

| **Radioisotope** | Lead-212 ($^{212}$Pb) | 10.6 hours | β−-emitter; in vivo generator of α-particles via $^{212}$Bi & $^{212}$Po | High LET (~100 keV/µm); Short range (50-100 µm) | Suitable for TAT; decay progeny deliver therapeutic dose 4 |

| Chelator | DOTAM (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic amide; TCMC) | N/A | N/A | N/A | Macrocyclic; superior to DOTA for $^{212}$Pb chelation; forms highly stable complex with $^{212}$Pb 3 |

| Targeting Ligand| GRPR1 Antagonist | N/A | N/A | N/A | Peptide antagonist; specifically targets GRPR; preferred over agonists for RLT 1 |

This table provides a concise summary of the fundamental building blocks of 212Pb-DOTAM-GRPR1, highlighting how their individual properties contribute to the overall design and intended function of this targeted alpha therapeutic.

III. Mechanism of Action

The therapeutic effect of 212Pb-DOTAM-GRPR1 is achieved through a multi-step process that begins with selective targeting and culminates in potent, localized cytotoxicity.

A. Selective Targeting of Gastrin-Releasing Peptide Receptor (GRPR)

Upon intravenous administration, the GRPR1 antagonist component of 212Pb-DOTAM-GRPR1 circulates through the bloodstream and selectively seeks out and binds to Gastrin-Releasing Peptide Receptors (GRPR) that are present on the surface of tumor cells.2 GRPR is a G protein-coupled receptor (GPCR) that exhibits significantly upregulated expression in a variety of cancer types when compared to most normal adult tissues.1 This differential expression provides a molecular basis for the targeted delivery of the radiopharmaceutical, aiming to concentrate the cytotoxic $^{212}$Pb payload predominantly at the site of malignancy while minimizing exposure and potential damage to healthy cells that exhibit low or negligible GRPR expression.2

### B. Delivery and Cytotoxic Effects of Alpha ($\alpha$) Particle Radiation

Once the 212Pb-DOTAM-GRPR1 conjugate is bound to GRPR on the tumor cell surface, the anchored $^{212}$Pb undergoes radioactive decay. This decay process leads to the in situ generation of its daughter radionuclides, notably $^{212}$Bi and $^{212}$Po, which are potent alpha emitters.7 These high-energy alpha particles are released in close proximity to, or within, the targeted tumor cell. A crucial characteristic of these alpha particles is their very short path length in biological tissue, typically ranging from 50 to 100 micrometers, which is equivalent to approximately 2 to 5 cell diameters.4 This extremely limited range ensures that the highly destructive energy of the alpha particles is deposited primarily within the targeted tumor cell and its immediate microenvironment. Consequently, this localized radiation delivery significantly minimizes the potential for collateral damage to surrounding healthy tissues that are beyond the alpha particles' reach.4 It is noteworthy that the efficacy of this targeted alpha therapy does not strictly require internalization of the receptor-ligand complex. Even if the GRPR1 antagonist remains bound to the cell surface, the emitted alpha particles possess sufficient energy and range to traverse cellular compartments and reach the cell nucleus to inflict damage.4

C. Induction of DNA Double-Strand Breaks and Tumor Cell Death

The therapeutic efficacy of alpha particles stems from their high linear energy transfer (LET). As alpha particles traverse cellular material, they create dense ionization tracks, depositing a large amount of energy in a very small volume. This concentrated energy deposition results in complex and, critically, difficult-to-repair DNA double-strand breaks (DSBs) within the nucleus of the targeted tumor cells.[4] DSBs are among the most lethal forms of DNA damage. The cell's capacity to accurately repair such extensive and clustered damage induced by high-LET radiation is limited. Consequently, these irreparable DSBs overwhelm cellular repair mechanisms, ultimately triggering programmed cell death (apoptosis) or necrotic cell death, leading to the elimination of the cancer cell.[6] The potency of this mechanism is remarkable, with estimates suggesting that fewer than five alpha particle traversals through a cell nucleus can be sufficient to induce cell kill.[4]

Furthermore, due to the physical range of alpha particles, there can be a "crossfire effect" where adjacent tumor cells that may not have directly bound the radiopharmaceutical can still be irradiated and killed if they are within the 50-100 µm radius of an alpha-emitting source.[7] This localized crossfire can contribute to a more comprehensive tumoricidal effect within a densely packed tumor. The direct and highly damaging nature of alpha particle-induced DNA lesions is also thought to hinder the development of acquired resistance mechanisms in tumor cells, a common challenge with many conventional cancer therapies.[4] This robust cell-killing mechanism suggests that 212Pb-DOTAM-GRPR1 could be particularly effective against tumors that have become refractory to other treatments which typically induce simpler, more reparable DNA damage.

IV. The Gastrin-Releasing Peptide Receptor (GRPR) as a Therapeutic Target

The choice of the Gastrin-Releasing Peptide Receptor (GRPR) as the molecular target for 212Pb-DOTAM-GRPR1 is based on its distinct biological roles and its pattern of expression in cancerous tissues.

A. Biological Functions and Signaling Pathways of GRPR

GRPR, also identified as Bombesin Receptor Subtype 2 (BB2), is a member of the G-protein coupled receptor (GPCR) superfamily, specifically within the bombesin receptor family.[1] The primary endogenous ligand for GRPR is gastrin-releasing peptide (GRP), a neuropeptide that plays a regulatory role in a multitude of physiological processes, particularly within the gastrointestinal tract and the central nervous system.[1] Normal functions mediated by GRP/GRPR signaling include the release of gastrointestinal hormones (e.g., gastrin), modulation of pancreatic and gastric exocrine secretions, and regulation of smooth muscle contraction.[9]

In the context of oncology, GRPR signaling pathways are frequently co-opted by cancer cells to promote their growth and survival. Activation of GRPR by GRP or related peptides can stimulate critical oncogenic processes, including cell proliferation, migration, angiogenesis (the formation of new blood vessels that supply tumors), and invasion into surrounding tissues.[2] Often, cancer cells produce GRP themselves, leading to an autocrine loop that perpetuates these growth-promoting signals.[9] Key intracellular signaling cascades activated downstream of GRPR include the PI3K/AKT pathway and the MAPK pathway, both of which are central to cell survival and proliferation.[20] Moreover, GRPR signaling has been implicated in promoting epithelial-to-mesenchymal transition (EMT). EMT is a cellular reprogramming process whereby epithelial cancer cells acquire mesenchymal characteristics, such as increased motility and invasiveness, which are crucial for the metastatic dissemination of cancer.[9] Thus, GRPR is not merely a passive docking site on cancer cells; its activation actively contributes to malignant progression. This dual role—a target for drug delivery and an active player in tumor biology—suggests that therapies targeting GRPR might not only deliver a cytotoxic payload but could also, by eliminating GRPR-expressing cells, disrupt these pro-tumorigenic signaling networks.

B. Overexpression of GRPR in Human Malignancies

A pivotal characteristic that makes GRPR an attractive target for cancer therapy is its significant and widespread overexpression in a diverse range of human malignancies when compared to its expression levels in most normal adult tissues.[1] This differential expression pattern provides a therapeutic window, allowing for targeted agents to preferentially accumulate in tumor tissues.

Notable cancers with high GRPR expression include:

  • Prostate Cancer: GRPR is highly expressed in a substantial percentage of prostate cancers, with reports indicating expression in 62-100% of cases, encompassing both primary tumors and metastatic lesions.[1] The correlation between GRPR expression and clinicopathological features like Gleason score or disease stage has yielded somewhat varied results across different studies, with some suggesting a positive correlation with higher-grade disease or relapse, while others report an inverse correlation or higher detection in lower-grade tumors.[20]
  • Breast Cancer: Overexpression is observed in 38-75.8% of breast cancers, with a particularly strong association with estrogen receptor (ER)-positive tumors.[1] Importantly, high GRPR expression in primary breast tumors often correlates with its presence in metastatic axillary lymph nodes.[22]
  • Lung Cancer: GRPR overexpression is noted in lung cancer, including 85-100% of small cell lung cancers (SCLC) and also in lung adenocarcinoma (LUAD).[1] In LUAD, higher GRPR expression has been associated with poorer overall survival outcomes and an increased propensity for metastasis.[23]
  • Cervical Cancer: Studies have demonstrated GRPR overexpression in a notable proportion of cervical cancers. For instance, one study found that 100% of primary adenocarcinomas and 63% of primary squamous cell carcinomas of the cervix overexpressed GRPR at an immunoreactive score (IRS) of 6 or higher, a threshold often used for patient selection in clinical trials.[7]
  • Colorectal Cancer: GRPR overexpression has been documented in colorectal malignancies.[1]
  • Melanoma: Cutaneous melanoma is another cancer type where GRPR overexpression has been observed.[1]
  • Other Cancers: The list of GRPR-expressing cancers extends to pancreatic cancer (approximately 75%), various brain tumors (including 85% of glioblastomas), head and neck cancers (up to 100%), and cancers of the uterus, ovary, and kidney.[9] While neuroendocrine tumors (NETs) can also express GRPR, other receptors like SSTR2 are more commonly targeted in NETs with different radioligand therapies.[8]

C. Clinical Significance of GRPR Expression

The overexpression of GRPR on tumor cells, juxtaposed with its generally lower expression in most healthy adult tissues, forms the cornerstone of its clinical significance as a therapeutic target.[2] This differential expression is anticipated to facilitate selective tumor targeting and thereby enhance the therapeutic ratio of GRPR-directed agents. However, it is important to recognize that GRPR expression can exhibit considerable heterogeneity, both between different tumor types and even within individual tumors or among patients with the same cancer type.[16] This variability underscores the critical need for reliable methods to assess GRPR status in patients to guide treatment decisions.

The development of GRPR-targeted PET imaging agents, such as those utilizing Gallium-68 (e.g., $^{68}$Ga-RM2, a GRPR antagonist), allows for non-invasive visualization and quantification of GRPR expression in tumors throughout the body.[20] Such imaging can play a crucial role in identifying patients whose tumors express sufficient levels of GRPR to be likely candidates for GRPR-targeted radioligand therapy like 212Pb-DOTAM-GRPR1. Indeed, tumor response to 212Pb-DOTAM-GRPR1 is expected to strongly correlate with the level of GRPR overexpression.[24]

In specific malignancies like prostate cancer, GRPR-targeted imaging and therapy may offer a complementary approach to those targeting other well-established markers like Prostate-Specific Membrane Antigen (PSMA). Emerging evidence suggests that some prostate tumors may have low or absent PSMA expression but still express GRPR, potentially identifying a patient subpopulation that could benefit from GRPR-targeted agents when PSMA-targeted options are less suitable.[20] The prognostic value of GRPR expression is also an area of active investigation, with some studies, such as in LUAD, linking higher GRPR levels to worse clinical outcomes [23], while findings in other cancers like breast cancer have been more complex and may depend on other co-expressed markers like ER status.[22] This highlights the necessity of companion diagnostics, whether imaging-based or immunohistochemical, to ensure that 212Pb-DOTAM-GRPR1 is administered to patients most likely to derive clinical benefit.

Table 2: GRPR Overexpression Across Various Cancer Types

Cancer TypeReported Overexpression Rates/LevelsKey Associations/NotesPrimary Source(s)
Prostate Cancer62-100%Primary and metastatic lesions; variable correlation with Gleason score; potential role in PSMA-low tumors9
Breast Cancer38-75.8%Strongly associated with Estrogen Receptor (ER) positivity; expression in primary often mirrors metastatic nodes9
Lung Cancer (SCLC)85-100%9
Lung Cancer (Adenocarcinoma)Elevated levelsAssociated with worse overall survival and metastasis23
Cervical Cancer (Adeno)100% (IRS $\geq$6 in one study)High expression threshold met for trial eligibility16
Cervical Cancer (Squamous)63% (IRS $\geq$6 in one study)High expression threshold met for trial eligibility16
Colorectal CancerOverexpressed1
MelanomaOverexpressed1
Pancreatic Cancer~75%9
Head and Neck CancerUp to 100%9
Brain Cancer (Glioblastoma)~85%9

This table summarizes the widespread overexpression of GRPR, reinforcing its potential as a broad-spectrum target for radioligand therapies like 212Pb-DOTAM-GRPR1.

V. Preclinical Evaluation of 212Pb-DOTAM-GRPR1

The transition of 212Pb-DOTAM-GRPR1 from a conceptual therapeutic to a clinical candidate was underpinned by a series of preclinical investigations designed to assess its biological activity, pharmacokinetic profile, efficacy, and safety in relevant in vitro and in vivo models.

A. In Vitro Studies: Receptor Affinity, Internalization, and Cytotoxicity

While the provided documentation does not offer exhaustive details on specific in vitro parameters for 212Pb-DOTAM-GRPR1, such as precise binding affinities (e.g., Kd​ values), internalization kinetics, or comprehensive cell viability assays (e.g., IC$_{50}$ values), the general expectations for such a compound can be inferred. Standard preclinical characterization would involve confirming the high-affinity and specific binding of the GRPR1 antagonist component (both unlabeled and radiolabeled) to GRPR-expressing cancer cell lines. Cellular uptake mechanisms, including the extent and rate of internalization of the receptor-ligand complex, would typically be assessed, although, as noted, internalization is not strictly essential for the efficacy of alpha-emitters. Dose-dependent cytotoxicity assays would be performed to quantify the ability of $^{212}$Pb-DOTAM-GRPR1 to kill GRPR-positive cancer cells while sparing GRPR-negative cells. For instance, studies with a different $^{212}$Pb-labeled agent targeting PSMA, ADVC001, demonstrated nanomolar binding affinities, efficient cellular internalization, and potent, specific cytotoxic activity against PSMA-positive cancer cell lines.[6] Similar rigorous in vitro validation would be anticipated for 212Pb-DOTAM-GRPR1 to establish its fundamental biological activity before advancing to animal models.

B. In Vivo Animal Models (Primarily GRPR-positive prostate cancer xenografts)

Preclinical in vivo studies, predominantly utilizing mouse xenograft models bearing GRPR-positive human prostate tumors, were crucial for evaluating the pharmacokinetics, biodistribution, dosimetry, and anti-tumor efficacy of 212Pb-DOTAM-GRPR1.[7]

Pharmacokinetics and Biodistribution:

These studies tracked the distribution and clearance of intravenously administered $^{212}$Pb-DOTAM-GRPR1 over time. Tumor targeting was observed, with uptake in GRPR-positive tumors reaching up to 5% of the injected dose per gram of tissue (%ID/g) at 24 hours post-injection.7 It is important to contextualize this uptake value; for example, a different $^{212}$Pb-labeled Bicycle Radionuclide Conjugate, $^{212}Pb−BCY20603targetingMT1−MMP,showedmuchhighertumoruptakeof>45Acriticalaspectofpreclinicaldevelopmentwastheoptimizationofthedrugformulation,particularlytheamountoftheGRPR1peptideadministered.Itwasfoundthatincreasingthepeptidemassinjected(from28ngto280ng)significantlyreducedoff−targetbindinganduptake,notablyinthepancreas(anorganwithphysiologicalGRPRexpression),byapproximately30∗∗TumorTargetingandRetention:∗∗TheradiopharmaceuticaldemonstratedpreferentialaccumulationinGRPR−expressingtumors,andthisaccumulationwasshowntocorrelatewiththelevelsofGRPRexpression.[2]TheantagonistnatureoftheGRPR1targetingligandisbelievedtocontributetofavorabletumorretentioncharacteristics,allowingforasustainedradiationdosetothetumorcells.[2,7]∗∗DosimetryandRadiationDosetoOrgans:∗∗Radiationdosimetrystudieswereconductedtoestimatetheabsorbedradiationdosestovariousorgansandthetumor.Thesestudiesconsistentlyidentifiedthekidneysastheprimarydose−limitingorgan.[7,11,15]Thisisacommonobservationformanypeptide−basedradiopharmaceuticals,astheyareoftenclearedfromthebodyviarenalexcretionandcanundergoreabsorptioninthekidneytubules.Encouragingly,thekidneyuptakeof[^{212}Pb]Pb−DOTAM−GRPR1wasreportedtobeconsiderablylowerthanthatobservedforsomeotherradiolabeledsomatostatinreceptorligands,suchas[^{212}Pb]Pb−DOTAMTATE,evenwhenthelatterwasadministeredwithrenalprotectiveagents.[11]Interestingly,attemptstoreducekidneyretentionof[^{212}$Pb]Pb-DOTAM-GRPR1 using standard renal blocking agents, such as positively charged amino acids or colchicine, were not successful in one preclinical study.11 This suggests that the mechanisms of renal uptake or retention for this specific GRPR antagonist conjugate might differ from those targeted by these conventional renal protectants, or that its baseline renal uptake is already sufficiently optimized. This finding, if translatable to humans, could simplify clinical administration protocols by potentially obviating the need for co-administered renal protective infusions.

C. Anti-Tumor Efficacy Studies

Preclinical efficacy studies provided compelling evidence of the anti-tumor activity of 212Pb-DOTAM-GRPR1. In animal models, typically mice bearing human prostate cancer xenografts with high GRPR expression, treatment with $^{212}$Pb-DOTAM-GRPR1 led to significant inhibition of tumor growth when compared to untreated control groups.[2] Specifically, in one key efficacy study, GRPR-positive prostate tumor-bearing mice that received a fractionated dosing regimen of four injections of 370 kBq of $^{212}$Pb-DOTAM-GRPR1 at 3-week intervals exhibited a median survival time of 19 weeks. This was a substantial improvement over the median survival of 9 weeks observed in the control group that received only a buffer solution.[7]

The investigation of fractionated dosing schedules was an important element of the preclinical program. Administering the total therapeutic dose in smaller, spaced-out fractions is a strategy often employed with potent cytotoxic agents, including alpha emitters, to manage potential toxicities while preserving or even enhancing therapeutic efficacy.[2] This approach aims to allow healthy tissues, particularly dose-limiting organs like the kidneys, time to recover between doses, while still delivering a cumulative lethal radiation dose to the more sensitive tumor cells. The success of such a regimen in preclinical models, as demonstrated by the survival benefit, supports its consideration for clinical trial designs. While direct comparisons are nuanced, the potent anti-tumor activity observed with other $^{212}$Pb-based agents, such as the complete tumor regressions seen with $^{212}$Pb-BCY20603 in a different tumor model [26], further underscores the high therapeutic potential inherent in $^{212}$Pb-based radiopharmaceuticals.

D. Safety and Toxicology Profile (including Maximum Tolerated Dose - MTD)

The preclinical safety and toxicology assessment of 212Pb-DOTAM-GRPR1 indicated that the compound exhibited an acceptable toxicity profile in animal models, provided that dosing was carefully controlled.2 Formal toxicity studies in mice were conducted to determine non-toxic dose levels and to identify potential dose-limiting toxicities. These studies revealed that a single intravenous injection of up to 1,665 kBq of $^{212}$Pb-DOTAM-GRPR1 was tolerated without overt signs of toxicity in mice.7 As previously mentioned, the kidneys were identified as the principal dose-limiting organ, consistent with the biodistribution and dosimetry findings.7 The establishment of a preclinical MTD and an understanding of the toxicity profile were essential prerequisites for advancing 212Pb-DOTAM-GRPR1 into human clinical trials.

### Table 3: Summary of Significant Preclinical Findings for 212Pb-DOTAM-GRPR1

| Parameter | Finding | Snippet Source(s) |

|-----------------------------------------------|---------------------------------------------------------------------------------------------------------|----------------------------|

| **Tumor Model** | GRPR-positive prostate tumor-bearing mice | 7 |

| **Tumor Uptake (%ID/g)** | Up to 5% at 24 hours | 7 |

| **Key Off-Target Organ (Uptake Reduction)** | Pancreas (uptake reduced ~30% by increasing peptide dose from 28 ng to 280 ng) | 7 |

| **Dose-Limiting Organ** | Kidney | 11 |

| **Non-Toxic Single Dose (Mice)** | Up to 1,665 kBq | 11 |

| **Efficacy (Survival Benefit vs. Control)** | Median survival 19 weeks vs. 9 weeks | 7 |

| **Efficacy (Dosing Regimen for Benefit)** | 4 injections of 370 kBq at 3-week intervals | 7 |

| **Renal Blocker Efficacy** | Positively charged amino acids or colchicine did not reduce kidney retention in one study | 11 |

This table concisely summarizes the key outcomes from the preclinical research program, which collectively provided the scientific rationale and safety justification for initiating clinical evaluation of 212Pb-DOTAM-GRPR1 in humans.

## VI. Clinical Development of 212Pb-DOTAM-GRPR1

The promising preclinical data for 212Pb-DOTAM-GRPR1, demonstrating tumor targeting, anti-tumor efficacy, and a manageable safety profile in animal models, provided a strong foundation for its advancement into clinical development.

### A. Overview of the Clinical Trial Program (Orano Med)

Orano Med is the pharmaceutical company sponsoring and leading the clinical translation of 212Pb-DOTAM-GRPR1.4 Reflecting their commitment to advancing targeted alpha therapies, Orano Med initiated a Phase I clinical trial for 212Pb-DOTAM-GRPR1 in early 2023.1 This first-in-human study was designed to evaluate the safety, tolerability, dosimetry, and preliminary efficacy of the radiopharmaceutical in patients diagnosed with advanced cancers that express the GRPR target.2 The decision to proceed to the clinical phase was directly supported by the comprehensive preclinical evidence package which demonstrated the safety, tolerability, and therapeutic potential of [$^{212}$Pb]Pb-DOTAM-GRPR1 in relevant xenograft models.7 A news release from Orano Med on January 4, 2023, formally announced the commencement of this Phase I trial, marking a significant milestone in the development of this novel alpha radioligand therapy.14

B. Phase I Clinical Trial (NCT05283330)

The pivotal first-in-human study for 212Pb-DOTAM-GRPR1 is registered under the identifier NCT05283330.

Study Title: The official title of the trial is "A Phase 1 Open-Label Dose Escalation and Expansion Study to Determine the Safety, Tolerability, Dosimetry, and Preliminary Efficacy of $^{212}$Pb-DOTAM-GRPR1 in Adult Subjects With Recurrent or Metastatic GRPR-expressing Tumors".[27]

Design, Objectives, and Key Endpoints:

The trial employs an open-label design, incorporating both dose escalation and dose expansion phases.27 The dose escalation portion follows a classic 3+3 design, a standard approach in Phase I oncology trials to determine the maximum tolerated dose (MTD).29 This involves enrolling cohorts of 3-6 patients at successively higher dose levels of $^{212}$Pb-DOTAM-GRPR1, encompassing both single ascending dose (SAD) and multiple ascending dose (MAD) regimens, until the MTD or recommended Phase 2 dose (RP2D) is identified.29

The primary objectives of this Phase I study are to:

  • Evaluate the safety profile and tolerability of $^{212}$Pb-DOTAM-GRPR1.
  • Determine the MTD and subsequently the RP2D for further clinical investigation.
  • Assess the radiation dosimetry, quantifying the absorbed radiation dose to tumors and critical normal organs.[2] Secondary and exploratory objectives include gathering preliminary data on the anti-tumor efficacy of $^{212}$Pb-DOTAM-GRPR1, which may involve assessing objective response rates, duration of response, and other measures of clinical benefit.[2] The inclusion of multiple GRPR-expressing tumor types within this single Phase I trial is characteristic of a "basket trial" design. This efficient strategy is well-suited for early-phase studies of targeted therapies, as it allows for the simultaneous assessment of safety and preliminary efficacy signals across different cancers that share the common molecular target (GRPR). This can accelerate enrollment and provide early indications of which tumor types may be most responsive, guiding the design of subsequent, more focused Phase II trials.

Patient Population and Target Indications:

The study enrolls adult subjects diagnosed with recurrent or metastatic, histologically confirmed solid tumors that express GRPR.27 Eligible patients are typically those whose disease has progressed despite receiving at least two prior lines of systemic therapy.29 A critical eligibility criterion is the confirmation of GRPR expression in tumor tissue by immunohistochemistry (IHC). Specific IHC criteria mentioned include requirements such as 51-80% of tumor cells staining positively with moderate intensity, or achieving an immunoreactive score (IRS) of 6 or higher.16 This stringent biomarker-driven patient selection is fundamental to precision oncology, aiming to enrich the trial population with patients most likely to benefit from the targeted therapy, thereby maximizing the chance of observing a drug effect and ensuring that only patients whose tumors express the target at a potentially therapeutic level are enrolled.

The target indications listed for this Phase I trial are broad, reflecting the widespread expression of GRPR, and include:

  • Breast Cancer
  • Colonic Cancer
  • A general category of GRPR-positive tumors
  • Cutaneous Malignant Melanoma
  • Metastatic Prostate Carcinoma
  • Non-Small Cell Lung Cancer (NSCLC)
  • Uterine Cervical Cancer.[16]

Current Status (as of latest snippets) and Investigational Sites:

As of the latest available information 27, the NCT05283330 trial is actively recruiting participants.27 The trial officially commenced on December 22, 2022.27 The sponsor of the trial is Orano Med LLC.27 Investigational sites for this study are located within the United States.27 One notable participating institution is the Markey Cancer Center at the University of Kentucky, which has a particular interest in evaluating the agent for cervical cancer, a disease with high incidence in the region.21

Preliminary Safety, Tolerability, and Dosimetry Data:

The provided research materials do not contain any specific preliminary clinical data (e.g., safety, tolerability, dosimetry, or efficacy results) from the ongoing NCT05283330 trial. As the trial is actively recruiting and in its early phase, such data would typically be presented at scientific conferences or published once sufficient patient cohorts have been evaluated and data analyzed. The cautious, stepwise approach of clinical development dictates that human safety must be thoroughly established in Phase I before efficacy can be robustly assessed in later phases, despite the promising preclinical efficacy signals.

Table 4: Details of the Phase I Clinical Trial NCT05283330

FeatureDetailsSource(s)
Trial IdentifierNCT0528333027
Full Title"A Phase 1 Open-Label Dose Escalation and Expansion Study to Determine the Safety, Tolerability, Dosimetry, and Preliminary Efficacy of $^{212}$Pb-DOTAM-GRPR1 in Adult Subjects With Recurrent or Metastatic GRPR-expressing Tumors"27
SponsorOrano Med LLC27
PhasePhase 127
StatusRecruiting (as of May 2025 or latest snippet)27
Start DateDecember 22, 202227
Primary ObjectivesDetermine safety, tolerability, Maximum Tolerated Dose (MTD), Recommended Phase 2 Dose (RP2D), and dosimetry27
Key IndicationsRecurrent/metastatic GRPR-expressing tumors, including: Breast, Colon, Melanoma, Prostate, Non-Small Cell Lung, and Cervical Cancers27
Study DesignOpen-label, dose escalation (Single Ascending Dose/Multiple Ascending Dose, classic 3+3 design) and dose expansion phases27
Key EligibilityAdult subjects; GRPR expression confirmed by Immunohistochemistry (IHC) (e.g., IRS $\geq$6 or 51-80% cells with moderate staining); progressed on prior therapies16
LocationsUnited States27

This table offers a structured overview of the pivotal first-in-human clinical trial for 212Pb-DOTAM-GRPR1, consolidating key administrative and scientific details essential for understanding its current development stage.

VII. Therapeutic Potential and Target Indications

The therapeutic potential of 212Pb-DOTAM-GRPR1 spans a range of solid tumors characterized by the overexpression of its molecular target, the Gastrin-Releasing Peptide Receptor. This potential is supported by the widespread expression of GRPR in various malignancies, encouraging preclinical data, and the inclusion of multiple cancer types in the ongoing Phase I clinical trial. The agent is primarily positioned to address unmet medical needs in patients with advanced, recurrent, or metastatic cancers that have become refractory to standard therapies.

A. Prostate Cancer

Prostate cancer exhibits high GRPR expression in a significant proportion of cases (62-100%).[1] Preclinical studies using prostate cancer models have demonstrated the anti-tumor efficacy of $^{212}$Pb-DOTAM-GRPR1.[2] Consequently, metastatic prostate carcinoma is one of the key indications in the Phase I trial.[27] A particularly interesting avenue for GRPR-targeted therapy in prostate cancer is its potential utility in tumors with low or absent expression of Prostate-Specific Membrane Antigen (PSMA).[20] As PSMA-targeted radioligand therapies (e.g., $^{177}$Lu-PSMA-617) become more established, a subset of patients with PSMA-negative disease will require alternative targeted approaches. If GRPR is upregulated in these PSMA-low tumors, 212Pb-DOTAM-GRPR1 could offer a valuable therapeutic option. Furthermore, GRPR is notably absent on salivary and lacrimal glands, which are sites of PSMA expression leading to xerostomia (dry mouth) as a common side effect of PSMA-targeted RLT. The lack of GRPR expression in these glands suggests that GRPR-targeted agents like 212Pb-DOTAM-GRPR1 might avoid this particular toxicity.[20]

B. Breast Cancer

GRPR is significantly overexpressed in breast cancer, particularly in estrogen receptor-positive (ER+) subtypes, with reported rates of 38-75.8%.[1] This makes breast cancer a strong candidate indication, and it is included in the Phase I trial.[27] Research suggests that GRPR targeting holds considerable promise for both imaging and treatment of ER-positive breast cancer.[22] For patients with metastatic or recurrent ER+ breast cancer who have progressed on endocrine therapies and chemotherapy, 212Pb-DOTAM-GRPR1 could represent a novel targeted approach if their tumors express GRPR.

C. Cervical Cancer

High levels of GRPR expression have been observed in cervical cancer, especially in adenocarcinomas (100% in one study meeting IRS $\geq$6 criteria) and a majority of squamous cell carcinomas (63% meeting IRS $\geq$6).[16] Uterine cervical cancer is an indication in the ongoing Phase I trial.[27] There is a recognized need for more effective treatments for persistent, recurrent, or metastatic cervical cancer, particularly to control occult metastatic disease.[21] 212Pb-DOTAM-GRPR1 is being investigated as a potential therapy to address this unmet need in GRPR-expressing cervical cancers.

D. Other GRPR-Expressing Solid Tumors

The broad expression of GRPR across various malignancies has led to the inclusion of several other tumor types in the Phase I trial for 212Pb-DOTAM-GRPR1:

  • Colorectal Cancer: GRPR expression has been noted [1], and it is an indication in the clinical trial.[27] For patients with refractory metastatic colorectal cancer, 212Pb-DOTAM-GRPR1 could offer a new therapeutic line if GRPR is expressed.
  • Lung Cancer (NSCLC & SCLC): GRPR expression is documented in both non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC).[1] NSCLC is specifically included in the Phase I trial.[27] In lung adenocarcinoma (a subtype of NSCLC), higher GRPR expression has been linked to a poorer prognosis [23], suggesting a potential role for GRPR-targeted therapy in aggressive disease.
  • Melanoma: Cutaneous malignant melanoma also exhibits GRPR expression [1] and is part of the Phase I trial.[27] For patients with metastatic melanoma refractory to immunotherapy or targeted therapies (like BRAF/MEK inhibitors), 212Pb-DOTAM-GRPR1 could be a future option for GRPR-positive cases.

The overarching strategy appears to target patient populations with significant unmet medical needs, typically those with "recurrent or metastatic" disease who have often "progressed on at least 2 prior systemic therapies".[27] While the current focus is on these advanced/refractory settings, should 212Pb-DOTAM-GRPR1 demonstrate exceptional efficacy and a favorable safety profile, future investigations might explore its utility in earlier lines of treatment or in combination regimens for selected GRPR-positive cancers. However, such considerations are speculative at this stage and depend heavily on forthcoming clinical data.

E. Potential Advantages and Comparison with Other Radioligand Therapies

212Pb-DOTAM-GRPR1 possesses several features that may offer advantages:

  • Alpha Emitter Potency: The use of $^{212}$Pb, an alpha emitter, provides high LET radiation, leading to potent and localized cell kill with the potential to overcome resistance mechanisms that affect other therapies.[4]
  • GRPR Target Specificity: As discussed, GRPR targeting may offer advantages in specific clinical scenarios, such as PSMA-low prostate cancer, and potentially a different side effect profile (e.g., avoidance of xerostomia) compared to PSMA-targeted agents.[20]
  • DOTAM Chelator Stability: The DOTAM chelator is reported to be superior to DOTA for complexing $^{212}$Pb, ensuring high stability of the radiopharmaceutical in vivo.[4]
  • Antagonist Ligand Properties: The use of a GRPR antagonist (GRPR1) is expected to confer benefits such as improved tumor retention and a more favorable biodistribution profile compared to agonist-based approaches.[2]

While the broad therapeutic potential across these cancers is evident, its realization will critically depend on the prevalence and level of GRPR expression within specific patient cohorts and the consistent application of reliable companion diagnostics to identify suitable candidates.

VIII. Manufacturing, Supply, and Developer Overview

The successful clinical development and potential future commercialization of 212Pb-DOTAM-GRPR1 are heavily reliant on robust manufacturing processes for clinical-grade $^{212}$Pb and the capabilities of its developer, Orano Med.

A. Production of Clinical-Grade $^{212}$Pb

Orano Med has developed and implemented specialized processes for the extraction and purification of $^{212}Pb.[5]ThisradioisotopeistypicallyderivedfromparentisotopessuchasThorium−228(^{228}$Th) through generator systems.[10] Recognizing the critical need for a reliable and scalable supply of this rare alpha-emitting radioisotope, Orano Med has made substantial investments in dedicated production facilities. These include the Domestic Production Unit located in Plano, Texas, USA, and the Maurice Tubiana Laboratory in France.[2] In a significant development, Orano Med announced the inauguration of the world's first industrial-scale manufacturing facility for $^{212}$Pb-based radiopharmaceuticals in June 2024, underscoring their commitment to meeting the demands for research, ongoing and future clinical trials, and eventual commercial supply.[14]

The relatively short physical half-life of $^{212}$Pb (10.6 hours) imposes stringent requirements on the efficiency of production, quality control, and logistics.[10] This often necessitates that the final radiolabeling step (conjugating $^{212}$Pb to the DOTAM-GRPR1 molecule) be performed at or near the clinical site of administration, or that highly efficient systems for rapid shipment of ready-to-inject doses are in place.[10]

B. Orano Med's Role and Capabilities in Targeted Alpha Therapy

Orano Med, a subsidiary of the global nuclear fuel cycle company Orano, is dedicated to the development of a new generation of targeted cancer therapies utilizing the unique therapeutic properties of $^{212}$Pb.[4] The company's strategy is twofold: to build a robust pipeline of innovative $^{212}$Pb-based therapeutic agents and to establish and maintain a secure and reliable supply chain for these drugs.[5]

Orano Med possesses comprehensive expertise spanning the entire lifecycle of $^{212}$Pb radiopharmaceuticals. This includes mastery of $^{212}$Pb chemistry, advanced conjugation technologies (leveraging, in part, proprietary antibody site-specific conjugation methods through its subsidiary Macrocyclics, which also manufactures chelators), and the capability to radiolabel $^{212}$Pb to a diverse array of biological targeting vectors, such as peptides and antibodies.[4] To support its research and development activities, Orano Med operates preclinical laboratories in both the United States (Plano, Texas) and France.[4]

Beyond 212Pb-DOTAM-GRPR1, Orano Med's pipeline features other promising $^{212}Pb−basedcandidates.Notably,AlphaMedix(^{212}$Pb-DOTAMTATE), which targets somatostatin receptor 2 (SSTR2) for the treatment of neuroendocrine tumors (NETs), is currently in Phase 2 clinical trials and has received Breakthrough Therapy Designation from the U.S. Food and Drug Administration (FDA).[4] This regulatory milestone for AlphaMedix, while for a different agent, reflects positively on the potential of Orano Med's $^{212}$Pb platform and may inform regulatory pathways for 212Pb-DOTAM-GRPR1, assuming similarly compelling clinical data emerge.

Orano Med's vertical integration, encompassing control over the $^{212}$Pb supply chain from parent isotope processing to the production of the final radiopharmaceutical, including chelator manufacturing via Macrocyclics [4], provides a significant strategic advantage. This model mitigates risks associated with external reliance for a critical and rare radioisotope, ensuring quality, quantity, and timely availability. Furthermore, Orano Med actively engages in strategic collaborations with other biotechnology and pharmaceutical companies (e.g., Molecular Partners, Bicycle Therapeutics, Sanofi) to access novel targeting moieties (such as DARPins, Bicycle peptides, antibodies) and expand the range of cancers addressable by their $^{212}$Pb platform technology.[14] These partnerships enable Orano Med to leverage external expertise in ligand development, thereby accelerating the diversification of its pipeline and enhancing the overall potential of its core $^{212}$Pb technology.

IX. Challenges, Future Directions, and Regulatory Considerations

Despite the significant promise of 212Pb-DOTAM-GRPR1 and targeted alpha therapy in general, several challenges must be addressed for successful clinical translation and broader application. Future research will focus on overcoming these hurdles and further optimizing treatment strategies.

A. Potential Challenges in Clinical Translation and Broader Application

  • Managing Renal Toxicity: As identified in preclinical studies, the kidneys are the dose-limiting organ for $^{212}Pb−DOTAM−GRPR1.[11,15]Whilethecompoundshowedapotentiallymorefavorablekidneyretentionprofilecomparedtosomeotherpeptideradiopharmaceuticalsevenwithoutrenalblockers[11],carefulmonitoringoflong−termrenalfunctioninhumanpatientswillbeessential.Theobservationthatstandardrenalprotectiveagents(likeaminoacidinfusionsorcolchicine)didnotreducekidneyuptakeof[^{212}$Pb]Pb-DOTAM-GRPR1 in preclinical models [11] implies that if renal toxicity becomes a significant concern in clinical trials, alternative mitigation strategies might need to be explored, or dose adjustments may be necessary.
  • Patient Selection and Biomarker Standardization: The efficacy of 212Pb-DOTAM-GRPR1 is contingent upon adequate GRPR expression in tumors. Ensuring accurate, reproducible, and standardized GRPR expression testing, whether through immunohistochemistry (IHC) or GRPR-targeted PET imaging, across different clinical sites will be crucial for appropriate patient selection.[16] The inherent heterogeneity of GRPR expression within and between tumors remains a challenge that necessitates robust biomarker strategies.[20]
  • Logistics of Short Half-Life Isotope: The 10.6-hour physical half-life of $^{212}$Pb presents logistical complexities.[10] It demands highly efficient, centralized manufacturing of the isotope, rapid formulation and quality control of the radiopharmaceutical, and swift distribution to clinical administration sites. Alternatively, the deployment of on-site or local $^{212}$Pb generators and radiolabeling capabilities would be required to manage these time constraints effectively.
  • Cost and Accessibility: Advanced and highly specialized therapies like TATs often involve significant costs associated with isotope production, drug manufacturing, specialized handling, and administration. These factors could potentially limit broader patient accessibility if the therapy becomes approved, requiring careful consideration of health economics.
  • Radiation Safety for Operators: The production of $^{212}$Pb from parent isotopes like $^{228}$Th or $^{224}$Ra involves handling radioactive materials that can pose exposure risks (e.g., from radon gas, a decay product, and gamma radiation) to personnel involved in the manufacturing process. Robust radiation safety protocols and appropriately shielded facilities are essential to manage these risks.[10]

B. Future Research: Combination Therapies, Patient Selection Biomarkers

Future research endeavors are likely to focus on several key areas to maximize the therapeutic potential of 212Pb-DOTAM-GRPR1:

  • Combination Therapies: Exploring the synergistic potential of 212Pb-DOTAM-GRPR1 in combination with other anti-cancer treatments is a logical next step. This could include immunotherapies (given that radiation can modulate the tumor microenvironment and immune responses, as suggested for another $^{212}$Pb-PSMA agent [6]), conventional chemotherapy, or inhibitors of DNA damage response pathways, with the aim of enhancing overall efficacy or overcoming potential resistance mechanisms.
  • Refining Patient Selection Biomarkers: Beyond qualitative GRPR expression, research may focus on quantitative GRPR imaging to establish precise expression thresholds that correlate with clinical response. The identification of additional predictive biomarkers, beyond GRPR status alone, could further refine patient selection and personalize treatment strategies. The development of a diagnostic companion using the same GRPR1 targeting peptide but labeled with a diagnostic isotope (e.g., $^{68}$Ga for PET or $^{203}$Pb for SPECT) would create a true theranostic pair. This would allow for highly accurate patient stratification by directly visualizing the expected biodistribution of the therapeutic agent, a direction implied by the success of other theranostic systems.[17] * **Optimizing Dosing Schedules:** Further investigation into optimal dosing regimens, including the frequency and number of cycles of fractionated therapy, will be important to maximize the therapeutic index.[2] * **Expanding to New Indications:** If 212Pb-DOTAM-GRPR1 demonstrates significant activity in the currently targeted cancers, its evaluation could be expanded to other malignancies known to express GRPR but not yet included in clinical trials. ### C. Regulatory Landscape for Alpha Emitters (Contextual) The regulatory environment for novel radiopharmaceuticals, including alpha emitters, is evolving. The FDA has demonstrated a willingness to utilize expedited review pathways, such as Breakthrough Therapy Designation (BTD), for promising agents that address serious conditions with unmet medical needs. The BTD granted to Orano Med's AlphaMedix ($^{212}$Pb-DOTAMTATE) is a positive indicator in this regard.[30] For 212Pb-DOTAM-GRPR1, a clear demonstration of a favorable risk-benefit profile, supported by robust safety and efficacy data from well-designed and adequately powered clinical trials, will be paramount for regulatory approval. Standardization of dosimetry methodologies for alpha particles, which may involve considerations of microdosimetry due to the stochastic nature of energy deposition from high-LET radiation [32], will also be important for regulatory evaluation and ensuring consistent dose reporting. Currently, no specific European Medicines Agency (EMA) designations for 212Pb-DOTAM-GRPR1 are mentioned in the available documentation. The manufacturing scalability and logistics, critical for ensuring widespread access post-approval, will also be under regulatory scrutiny. Orano Med's significant investments in $^{212}$Pb production infrastructure [5] are proactive steps in addressing these long-term requirements.

The expertise developed by Orano Med with $^{212}$Pb and the DOTAM chelator effectively constitutes a platform technology.[4] By substituting the GRPR1 targeting peptide with other specific ligands (peptides, antibodies, DARPins, Bicycle peptides, etc.), this platform can be adapted to target a wide array of different cancer-associated molecular markers. This adaptability is evident in their development of AlphaMedix (targeting SSTR2) and their numerous collaborations aimed at identifying new targeting vectors for their $^{212}$Pb payload.[14] This platform approach allows Orano Med to leverage its core competencies in $^{212}$Pb production and radiochemistry across a diversified portfolio of oncology indications.

X. Conclusion: Synthesizing the Promise of 212Pb-DOTAM-GRPR1

A. Summary of Key Attributes and Therapeutic Rationale

212Pb-DOTAM-GRPR1 is an investigational radiopharmaceutical that embodies a sophisticated approach to cancer treatment through targeted alpha therapy. Its design integrates a potent alpha-emitting payload, $^{212}$Pb, with a highly specific targeting mechanism directed against the Gastrin-Releasing Peptide Receptor (GRPR), a protein frequently overexpressed on the surface of various cancer cells. The therapeutic rationale is compelling: the GRPR1 antagonist component ensures selective delivery to tumor sites; the stable DOTAM chelator securely sequesters the $^{212}$Pb; and upon decay, the radioisotope generates high-energy alpha particles that inflict lethal, localized radiation damage to cancer cells. Key advantages of this modality include the profound cytotoxicity of alpha particles, their short range in tissue which minimizes collateral damage to healthy bystander cells, and the potential to overcome resistance mechanisms that plague other therapies. Preclinical studies have provided a strong foundation, demonstrating encouraging anti-tumor efficacy and a manageable safety profile in relevant animal models, which collectively justified the initiation of human clinical trials.

B. Outlook for 212Pb-DOTAM-GRPR1 in Oncology

The ongoing Phase I clinical trial (NCT05283330) represents a critical juncture in the development of 212Pb-DOTAM-GRPR1. The outcomes of this trial will be instrumental in validating its safety, determining appropriate dosing for further studies, and providing initial insights into its efficacy in human patients with GRPR-expressing cancers. If successful, 212Pb-DOTAM-GRPR1 holds the potential to address significant unmet medical needs across a spectrum of advanced malignancies, particularly for patients whose tumors overexpress GRPR and have become refractory to existing standard-of-care treatments.

The development of 212Pb-DOTAM-GRPR1 is a clear manifestation of the principles of precision oncology: it involves the identification of a specific molecular abnormality in cancer cells (GRPR overexpression), the design of a drug (the radioligand) to selectively interact with that target, the delivery of a highly potent therapeutic mechanism (alpha particle radiation) to eradicate those targeted cells, and the use of biomarkers (GRPR expression levels) to guide patient selection. This tailored approach aims to maximize therapeutic benefit while minimizing systemic toxicity.

Furthermore, the clinical progression of 212Pb-DOTAM-GRPR1, alongside other $^{212}$Pb-based agents such as AlphaMedix, contributes significantly to the broader maturation of the targeted alpha therapy field. Each agent that advances through clinical development adds invaluable data and experience regarding the manufacturing, dosimetry, safety, efficacy, and practical application of this potent class of therapeutics. The collective knowledge gained will undoubtedly inform future drug design, refine clinical trial methodologies, and shape the regulatory landscape for TATs. The ultimate success of 212Pb-DOTAM-GRPR1 will depend on continued positive clinical trial results, the ability to effectively select patients who will benefit most, the ongoing management of any potential toxicities, and the scalability of manufacturing and supply to meet potential clinical demand. Should these aspects align favorably, 212Pb-DOTAM-GRPR1 could emerge as an important new therapeutic option in the oncologist's armamentarium.

Works cited

  1. Pb-212-GRPR - Oncidium Foundation, accessed June 9, 2025, https://www.oncidiumfoundation.org/molecule/pb-212-grpr/
  2. Lead-212 DOTAM-GRPR1: A Novel Radioimmunoconjugate for Targeted Cancer Therapy, accessed June 9, 2025, https://openmedscience.com/lead-212-dotam-grpr1-a-novel-radioimmunoconjugate-for-targeted-cancer-therapy/
  3. Definition of lead Pb 212-DOTAM-GRPR1 - NCI Drug Dictionary - NCI, accessed June 9, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/lead-pb-212-dotam-grpr1
  4. Targeted Alpha Therapy - Orano Med, accessed June 9, 2025, https://www.oranomed.com/en/targeted-alpha-therapy
  5. Our mission | Orano Med, accessed June 9, 2025, https://www.oranomed.com/en/about-us/our-ambition
  6. Abstract 588: Preclinical evaluation of a novel 212Pb-based, PSMA-targeted radioligand, 212Pb-ADVC001, for prostate cancer treatment - AACR Journals, accessed June 9, 2025, https://aacrjournals.org/cancerres/article/85/8_Supplement_1/588/754817/Abstract-588-Preclinical-evaluation-of-a-novel
  7. Preclinical Investigation of [212Pb] Pb-DOTAM-GRPR1 for Peptide ..., accessed June 9, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11533912/
  8. GRPR-targeting radiotheranostics for breast cancer management - PMC - PubMed Central, accessed June 9, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10657217/
  9. Gastrin-Releasing Peptide Receptor (GRPr) Promotes EMT, Growth ..., accessed June 9, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5867904/
  10. An Experimental Generator for Production of High-Purity 212Pb for ..., accessed June 9, 2025, https://jnm.snmjournals.org/content/64/1/173
  11. Preclinical Investigation of 212 Pb-DOTAMTATE for Peptide Receptor Radionuclide Therapy in a Neuroendocrine Tumor Model | Request PDF - ResearchGate, accessed June 9, 2025, https://www.researchgate.net/publication/333208077_Preclinical_Investigation_of_212_Pb-DOTAMTATE_for_Peptide_Receptor_Radionuclide_Therapy_in_a_Neuroendocrine_Tumor_Model
  12. Side-by-Side Comparison of the In Vivo Performance of [ 212 Pb]Pb-DOTAMTATE and Other SSTR2-Targeting Compounds - Journal of Nuclear Medicine, accessed June 9, 2025, https://jnm.snmjournals.org/content/early/2025/01/30/jnumed.124.268345
  13. Home - Orano Med, accessed June 9, 2025, https://www.oranomed.com/en
  14. All our News - Orano Med, accessed June 9, 2025, https://www.oranomed.com/en/orano-med-ressources/all-our-news
  15. Preclinical Investigation of 212Pb Pb-DOTAM-GRPR1 for Peptide Receptor Radionuclide Therapy in a Prostate Tumor Model - Orano Med, accessed June 9, 2025, https://www.oranomed.com/en/orano-med-ressources/publications/publications/preclinical-investigation-of-pb-pb-dotam-grpr1-for-peptide-receptor-radionuclide-therapy-in-a-prostate-tumor-model
  16. Human gastrin- releasing peptide receptor expression in women with uterine cervix cancer, accessed June 9, 2025, https://www.researchgate.net/publication/367393191_Human_gastrin-_releasing_peptide_receptor_expression_in_women_with_uterine_cervix_cancer
  17. Theranostic Perspectives in Prostate Cancer with the Gastrin-Releasing Peptide Receptor Antagonist NeoBOMB1: Preclinical and First Clinical Results | Journal of Nuclear Medicine, accessed June 9, 2025, https://jnm.snmjournals.org/content/58/1/75
  18. The chemical structure of the GRPR-targeting antagonist peptide... | Download Scientific Diagram - ResearchGate, accessed June 9, 2025, https://www.researchgate.net/figure/The-chemical-structure-of-the-GRPR-targeting-antagonist-peptide_fig8_391571629
  19. Gastrin-releasing peptide receptor (GRPR) mediates chemotaxis in neutrophils | PNAS, accessed June 9, 2025, https://www.pnas.org/doi/10.1073/pnas.1110996109
  20. Imaging and Therapy for Prostate-Specific Membrane Antigen (PSMA) Low-Expressing Prostate Cancers; Could Gastrin-Releasing Peptide Receptor (GRPR) Be the Next Target? - UroToday, accessed June 9, 2025, https://www.urotoday.com/clinical-trials/from-the-editor/145883-imaging-and-therapy-for-prostate-specific-membrane-antigen-psma-low-expressing-prostate-cancers-could-gastrin-releasing-peptide-receptor-grpr-be-the-next-target.html
  21. Markey study shows potential for new radiopharmaceutical cancer treatment, accessed June 9, 2025, https://medicine.uky.edu/news/markey-study-shows-potential-new-2023-02-24t09-13-25
  22. Expression of Gastrin-Releasing Peptide Receptor in Breast Cancer and Its Association with Pathologic, Biologic, and Clinical Parameters - PubMed, accessed June 9, 2025, https://pubmed.ncbi.nlm.nih.gov/28280221/
  23. GRPR Drives Metastasis via CRABP2 and FNDC4 Pathways in Lung Adenocarcinoma, accessed June 9, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11674891/
  24. Human gastrin- releasing peptide receptor expression in women with uterine cervix cancer, accessed June 9, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9905715/
  25. Comparison of 68Ga-GRPR PET/CT with 68Ga-PSMA PET/CT in initial diagnosing prostate cancer using histopathology: results from 207 participants | Journal of Nuclear Medicine, accessed June 9, 2025, https://jnm.snmjournals.org/content/63/supplement_2/2540
  26. Potent anti-tumor activity of a Lead-212 labelled MT1-MMP targeting Bicycle Radionuclide ConjugateTM, accessed June 9, 2025, https://www.bicycletherapeutics.com/wp-content/uploads/2023/05/Potent_anti-tumor_activity_of_a_Lead-212_labelled_MT1-MMP_targeting_Bicycle_Radionuclide_Conjugate.pdf
  27. 212Pb-DOTAM-GRPR1 - Drug Targets, Indications, Patents - Synapse, accessed June 9, 2025, https://synapse.patsnap.com/drug/d0785337a3eb44cb9e3b93fc5252bc4e
  28. 212Pb-DOTAM-GRPR1 - MedPath, accessed June 9, 2025, https://trial.medpath.com/drug/686aef1f101d7299/212pb-dotam-grpr1
  29. Safety and Tolerability of ²¹²Pb-DOTAM-GRPR1 in Adult Subjects With Recurrent or Metastatic GRPR-expressing Tumors - NCI, accessed June 9, 2025, https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCI-2022-07733
  30. FDA Grants Breakthrough Therapy Designation to Novel Agent in GEP-NETs, accessed June 9, 2025, https://www.targetedonc.com/view/fda-grants-breakthrough-therapy-designation-to-novel-agent-in-gep-nets
  31. RadioMedix and Orano Med receive FDA Breakthrough Therapy Designation for AlphaMedixTM in gastroenteropancreatic neuroendocrine tumors - Business Wire, accessed June 9, 2025, https://www.businesswire.com/news/home/20240212199538/en/RadioMedix-and-Orano-Med-receive-FDA-Breakthrough-Therapy-Designation-for-AlphaMedixTM-in-gastroenteropancreatic-neuroendocrine-tumors
  32. MIRD Pamphlet No. 22 (Abridged): Radiobiology and Dosimetry of α-Particle Emitters for Targeted Radionuclide Therapy | Journal of Nuclear Medicine, accessed June 9, 2025, https://jnm.snmjournals.org/content/51/2/311

Published at: June 9, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.