MedPath

Dalotuzumab Advanced Drug Monograph

Published:May 2, 2025

Generic Name

Dalotuzumab

Drug Type

Biotech

CAS Number

1005389-60-5

Dalotuzumab (MK-0646): A Comprehensive Review of its Development and Clinical Evaluation in Oncology

1. Executive Summary

Dalotuzumab, also known by its development code MK-0646, is an investigational, humanized IgG1 monoclonal antibody developed by Merck & Co., Inc., under license from Pierre Fabre SA.[1] Designed as a targeted therapy for various cancers, Dalotuzumab specifically binds to the Insulin-like Growth Factor 1 Receptor (IGF-1R), a key component of a signaling pathway implicated in cancer cell proliferation, survival, and resistance to therapy.[1] By inhibiting IGF-1R activation by its ligands (IGF-1 and IGF-2), Dalotuzumab aimed to block downstream signaling cascades, induce apoptosis, and potentially elicit antibody-dependent cellular cytotoxicity.[1]

The clinical development program for Dalotuzumab was extensive, encompassing numerous Phase I, II, and III trials across a spectrum of malignancies, including metastatic colorectal cancer (mCRC), non-small cell lung cancer (NSCLC), breast cancer, pancreatic cancer, small cell lung cancer (SCLC), and multiple myeloma.[1] It was evaluated both as a monotherapy and, more frequently, in combination with standard-of-care treatments such as chemotherapy (e.g., pemetrexed/cisplatin, irinotecan), EGFR inhibitors (e.g., cetuximab, erlotinib), and inhibitors of the PI3K/Akt/mTOR pathway (e.g., ridaforolimus).[5]

Despite a strong preclinical rationale and generally acceptable tolerability profile in clinical studies, Dalotuzumab consistently failed to demonstrate significant improvements in primary efficacy endpoints, such as progression-free survival (PFS) and overall survival (OS), when added to standard therapies in major randomized trials.[14] While the drug was often feasible to administer in combination regimens, hyperglycemia emerged as a characteristic and frequent adverse event, reflecting on-target engagement but also posing a management challenge.[1] Other toxicities were often related to the combination agents used.

Evidence strongly suggests that the clinical development of Dalotuzumab has been discontinued. This includes the premature termination of a pivotal Phase II/III trial in mCRC due to futility [18], the termination of other trials [16], and the absence of MK-0646 from recent Merck pipeline reports compared to its presence in earlier ones.[24] This outcome mirrors the broader difficulties encountered in targeting the IGF-1R pathway, where multiple drug candidates have faced similar setbacks.[26] The consistent failure across diverse tumor types and combination strategies points towards fundamental challenges, likely related to the complexity of the IGF signaling network, pathway redundancy, intricate feedback mechanisms, and the lack of robust predictive biomarkers to identify patient populations most likely to benefit, rather than issues confined to specific trial designs or indications.[14]

2. Dalotuzumab: Profile and Mechanism of Action

2.1 Drug Identification and Classification

Dalotuzumab is identified by several names and codes:

  • International Nonproprietary Name (INN): Dalotuzumab [28]
  • Development Codes: MK-0646, h7C10 [2]
  • DrugBank ID: DB11840 [7]
  • CAS Number: 1005389-60-5 [1]
  • FDA UNII: 6YI1L648RH [1]

Dalotuzumab is classified as a biotech therapeutic, specifically a protein-based therapy.[7] It is a humanized IgG1 kappa monoclonal antibody [1], derived from the murine monoclonal antibody 7C10.[2] The drug was developed by Merck & Co., Inc., under license from the French pharmaceutical company Pierre Fabre SA.[1] Its molecular formula is C6528H10086N1730O2018S40, with a calculated molar mass of approximately 146.4 kDa.[1] Detailed protein sequence information is publicly available.[28]

2.2 The Insulin-like Growth Factor (IGF) System and IGF-1R Pathway in Cancer

The IGF system plays a pivotal role in normal physiology but is also critically involved in the pathogenesis of numerous cancers.[1] This complex system, often referred to as the IGF axis, comprises ligands (Insulin, IGF-1, IGF-2), cell surface receptors (Insulin Receptor, IGF-1 Receptor, IGF-2 Receptor), and a family of seven high-affinity IGF binding proteins (IGFBP1-7) that modulate ligand availability.[1]

The IGF-1R is the primary signaling receptor within this axis.[1] It is a receptor tyrosine kinase (RTK) possessing a heterotetrameric structure, composed of two extracellular α-subunits that bind the ligands and two transmembrane β-subunits containing the intracellular tyrosine kinase domain.[1] Ligand binding (primarily IGF-1 or IGF-2) induces conformational changes, leading to receptor autophosphorylation and activation of the kinase domain. This initiates downstream signaling cascades primarily through the phosphorylation of adaptor proteins like Insulin Receptor Substrate (IRS) proteins (especially IRS-1) and Src-homology collagen (Shc).[1] Activation of IRS proteins predominantly triggers the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway, while Shc activation typically engages the Ras/Raf/MEK/Erk (MAPK) pathway.[1]

These signaling pathways are fundamental regulators of critical cellular processes, including cell survival, proliferation, differentiation, metabolism, and cell cycle progression.[1] Aberrant activation of the IGF-1R pathway is frequently observed in cancer. Overexpression or amplification of IGF-1R has been documented in various human malignancies, including NSCLC, breast cancer, colorectal cancer, and pancreatic cancer, and is often associated with tumor formation, increased metastatic potential, resistance to therapy, and worse patient prognosis.[1] Furthermore, elevated circulating levels of IGF-1 have been linked epidemiologically to an increased risk of developing certain cancers (e.g., prostate, colorectal) and poorer outcomes.[26] The critical role of IGF-1R signaling in driving cancer cell growth and survival provided a strong rationale for developing therapies targeting this receptor.[1]

2.3 Dalotuzumab's Mechanism of Action

Dalotuzumab was designed as a selective antagonist of IGF-1R signaling.[10] Its mechanism of action involves several key steps:

  • Target Binding: Dalotuzumab binds with high affinity (Kd reported as ~1 nM [39] or 30.3 pM) to the extracellular domain of the human IGF-1R (also known as CD221).[2]
  • Ligand Blockade: This binding effectively prevents the natural ligands, IGF-1 and IGF-2, from accessing their binding sites on the receptor.[1]
  • Inhibition of Receptor Activation: By blocking ligand binding, Dalotuzumab inhibits ligand-induced receptor autophosphorylation and activation.[2] This prevents the initiation of downstream signaling.
  • Downstream Pathway Inhibition: Consequently, Dalotuzumab inhibits the phosphorylation and activation of key downstream signaling molecules, notably Akt and IRS-1, thereby blocking signal transmission through the PI3K/Akt pathway.[2] Pharmacodynamic studies in Phase I confirmed inhibition of IGF-1R pathway signaling in treated tumors.[20]
  • Cellular Consequences: The blockade of IGF-1R signaling by Dalotuzumab leads to inhibition of tumor cell proliferation and induction of apoptosis and cell cycle arrest in preclinical models.[2]
  • Receptor Downregulation: Dalotuzumab has also been shown to induce the internalization and subsequent degradation of the IGF-1R, further reducing the potential for signaling.[13]
  • Immune Effector Function: As a humanized antibody of the IgG1 isotype, Dalotuzumab possesses the potential structural capability to mediate Antibody-Dependent Cellular Cytotoxicity (ADCC).[5] In vitro studies showed an increase in NK cell-mediated lysis of cancer cells in the presence of Dalotuzumab [36], suggesting this could be a secondary mechanism contributing to its potential anti-tumor effect.[6]

A key feature of Dalotuzumab is its high specificity for IGF-1R, with no reported cross-reactivity with the closely related Insulin Receptor (IR).[5] This specificity was likely designed to minimize the metabolic side effects, such as severe hypoglycemia, that could arise from inhibiting insulin signaling. However, the complexity of the IGF/insulin signaling network, including the existence of IR homodimers, IGF-1R homodimers, and IGF-1R/IR hybrid receptors, means that this specificity might inadvertently allow for compensatory signaling.[26] Cancer cells could potentially bypass the IGF-1R blockade by utilizing signaling through IR homodimers or hybrid receptors, which Dalotuzumab does not target. This inherent limitation, stemming from its designed specificity, may represent a fundamental reason for its lack of clinical efficacy despite effective engagement with IGF-1R.[26]

Furthermore, while Dalotuzumab demonstrated multiple relevant anti-cancer activities in preclinical settings (signal blockade, apoptosis induction, potential ADCC) [2], its failure in clinical trials suggests these mechanisms were insufficient to overcome the robustness of cancer signaling networks in vivo. The complex interplay within the tumor microenvironment, along with intrinsic or acquired resistance mechanisms such as pathway redundancy and feedback loop activation (e.g., involving PI3K/Akt/mTOR or Notch pathways), likely predominated over the anti-tumor effects of targeting IGF-1R alone.[15]

3. Clinical Development Program in Oncology

3.1 Overview

Dalotuzumab underwent extensive clinical evaluation, primarily in Phase I and Phase II studies, targeting a wide array of malignancies.[1] The program investigated Dalotuzumab both as a monotherapy and, more commonly, in combination with various standard-of-care agents, including chemotherapy, targeted therapies (EGFR inhibitors, mTOR inhibitors), and other investigational agents (AKT inhibitors, Notch inhibitors). Key indications explored included metastatic colorectal cancer (mCRC), non-small cell lung cancer (NSCLC), breast cancer, pancreatic cancer, small cell lung cancer (SCLC), multiple myeloma, and neuroendocrine tumors.[1] Despite the breadth of investigation, the program ultimately failed to demonstrate sufficient efficacy to support regulatory approval.

3.2 Key Clinical Trials Summary

The following table summarizes key clinical trials involving Dalotuzumab, highlighting the range of indications, combinations, and general outcomes based on the available information.

NCT ID / EudraCT ID / Other IDPhaseIndication(s)Combination TherapyStatusKey Efficacy FindingKey Safety FindingSource(s)
NCT00614393II/IIImCRC (KRAS WT, Chemorefractory)Cetuximab + IrinotecanTerminatedNo improvement in PFS/OS vs control; FutilityFeasible; Increased Gr≥3 Hyperglycemia, Asthenia18
NCT00925015ImCRC (Advanced)Cetuximab + IrinotecanCompletedSafety/PK lead-in for NCT00614393Tolerable (non-Japanese)6
(Japanese Study)ImCRC (Advanced, Japanese)Cetuximab + IrinotecanCompletedSafety/PK assessmentTolerable; Minimal PK interaction6
NCT01609231 / EUCTR2012-000317-36IIAmRC (Metastatic Rectal Cancer, High IGF-1/Low IGF-2)Irinotecan (vs Cetuximab + Irinotecan)TerminatedN/A (Terminated)N/A (Terminated)16
NCT00654420I/IINSCLC (Advanced, Refractory)ErlotinibCompletedNo improvement in PFS/OS/ORR vs Erlotinib aloneTolerable; Increased Hyperglycemia9
NCT00729742INSCLCErlotinibCompletedImaging studyN/A (Details limited)43
NCT00799240IINSCLC (Non-Squamous, Stage IV, Untreated)Pemetrexed + CisplatinCompletedSimilar ORR vs control; No clear benefitTolerable; Increased Gr≥3 Hyperglycemia4
NCIC CTG IND.190ISCLC (Extensive Stage, Untreated)Cisplatin + EtoposideCompletedORR 67%; RP2D determined (10 mg/kg weekly)Feasible; Increased dose-dependent Hyperglycemia17
NCT01605396IIBreast Cancer (ER+, High Ki67, Advanced)Ridaforolimus + ExemestaneCompletedNo improvement in PFS vs Ridaforolimus + Exemestane alone (trend favoring control)Tolerable (less stomatitis/pneumonitis than control)8
NCT00759785IBreast CancerMonotherapy or CombinationCompletedN/A (Details limited)N/A (Details limited)42
NCT00694356IAdvanced Solid TumorsMonotherapyCompletedSome clinical activity (SD, PRs); PD effects confirmedGenerally well-tolerated; Hyperglycemia (19%)3
NCT01233168IAdvanced Solid Tumors (Expansion in Ovarian/CRC)MK-2206 (AKT inh) or Ridaforolimus (mTOR inh) or MK-0752 (Notch inh)CompletedLimited efficacy (7 patients > 4 cycles)Tolerable with DLTs observed (Neutropenia, Rash, GI)39
(Pancreatic Study)I/IIPancreatic Cancer (Metastatic)Gemcitabine +/- ErlotinibCompletedImproved OS but not PFS vs Gemcitabine + ErlotinibTolerable; MTD established16
NCT00730379IAdvanced CancerRidaforolimus (mTOR inh)CompletedSafety/PK assessmentN/A (Details limited)11

Abbreviations: mCRC=Metastatic Colorectal Cancer; mRC=Metastatic Rectal Cancer; NSCLC=Non-Small Cell Lung Cancer; SCLC=Small Cell Lung Cancer; ER+=Estrogen Receptor Positive; KRAS WT=KRAS Wild-Type; PFS=Progression-Free Survival; OS=Overall Survival; ORR=Objective Response Rate; RP2D=Recommended Phase 2 Dose; PD=Pharmacodynamic; PK=Pharmacokinetic; Gr≥3=Grade 3 or higher; GI=Gastrointestinal; DLT=Dose-Limiting Toxicity; MTD=Maximum Tolerated Dose; N/A=Not Available.

3.3 Detailed Review by Indication

3.3.1 Metastatic Colorectal Cancer (mCRC)

The most significant trial in mCRC was the randomized, double-blind, Phase II/III study (NCT00614393) evaluating Dalotuzumab combined with cetuximab and irinotecan in patients with KRAS wild-type, chemorefractory disease.[18] This trial aimed to leverage the potential synergy between IGF-1R inhibition and EGFR inhibition, given preclinical evidence of pathway cross-talk.[6] Patients were randomized to receive cetuximab/irinotecan plus either weekly Dalotuzumab (10 mg/kg), bi-weekly Dalotuzumab (7.5 mg/kg after a loading dose), or placebo.[18] However, the trial was stopped prematurely for futility after an interim analysis of 344 patients showed no improvement in the primary endpoints of PFS or OS in either Dalotuzumab arm compared to placebo.[18] Median PFS was 3.9 months (weekly Dalotuzumab) and 5.4 months (bi-weekly Dalotuzumab) versus 5.6 months for placebo. Median OS was 10.8 months (weekly) and 11.6 months (bi-weekly) versus 14.0 months for placebo, indicating a negative trend for the Dalotuzumab arms.[18] Disease control rate was significantly lower in the weekly Dalotuzumab arm compared to placebo (42.3% vs 65.7%).[18] While the combination was deemed feasible, Grade 3 or higher hyperglycemia and asthenia were notably more frequent in the Dalotuzumab arms.[18] Exploratory biomarker analysis suggested high tumor IGF-1 mRNA levels might predict benefit from Dalotuzumab but resistance to cetuximab, hinting at complex interactions and the need for patient selection.[18] A Phase I safety/PK lead-in study (NCT00925015) and a similar Phase I study in Japanese patients established the tolerability of the triple combination prior to the Phase II/III trial.[6] A Phase IIA trial (NCT01609231 / EUCTR2012-000317-36) specifically targeting metastatic rectal cancer patients with a high IGF-1/low IGF-2 biomarker signature was initiated but subsequently terminated.[16]

3.3.2 Non-Small Cell Lung Cancer (NSCLC)

Dalotuzumab was investigated in NSCLC based on IGF-1R overexpression and its association with poor prognosis, particularly in adenocarcinoma and never-smokers.[4] A Phase I/II trial (NCT00654420) combined Dalotuzumab with the EGFR inhibitor erlotinib in patients with refractory advanced NSCLC.[9] The Phase I portion established Dalotuzumab 10 mg/kg weekly plus erlotinib 150 mg daily as safe.[14] However, the randomized Phase II part showed no significant improvement in PFS (2.5 vs 1.6 months), OS (6.6 vs 10.2 months), or ORR (2.7% vs 7.9%) for the combination compared to erlotinib alone.[14] Again, a trend towards worse OS was observed in the combination arm. The combination was tolerable, with hyperglycemia being a notable AE alongside typical erlotinib toxicities.[14]

Another Phase II trial (NCT00799240) evaluated Dalotuzumab added to first-line pemetrexed and cisplatin chemotherapy in patients with Stage IV non-squamous NSCLC.[4] This trial also failed to show a benefit, with similar response rates observed in both the combination and chemotherapy-alone arms.[22] The combination arm experienced a higher incidence of Grade 3-4 hyperglycemia (4 cases Gr 3, 1 case Gr 4) compared to the control arm.[4] The investigators concluded that predictive biomarkers would be essential for any further study in this population.[22] A Phase I imaging study (NCT00729742) also combined Dalotuzumab and erlotinib, but specific results are limited.[43]

Dalotuzumab was also tested in extensive-stage small-cell lung cancer (SCLC) in a Phase I trial (NCIC CTG IND.190) combined with standard cisplatin and etoposide.[17] The combination was feasible, with Dalotuzumab 10 mg/kg weekly determined as the recommended Phase II dose. An objective response rate of 67% was observed, but toxicities were consistent with chemotherapy plus dose-dependent hyperglycemia.[17]

3.3.3 Breast Cancer

In breast cancer, particularly ER-positive disease, aberrant PI3K/Akt/mTOR signaling is a key mechanism of endocrine resistance.[15] Inhibition of mTOR can lead to feedback upregulation of IGF-1R/IRS1 signaling, activating Akt and limiting therapeutic efficacy.[15] Combining an mTOR inhibitor with an IGF-1R inhibitor like Dalotuzumab was hypothesized to abrogate this feedback loop and enhance anti-tumor activity.[15] This led to a Phase II trial (NCT01605396) comparing ridaforolimus plus exemestane (R/E) with ridaforolimus plus Dalotuzumab plus exemestane (R/D/E) in postmenopausal women with advanced ER+, high-proliferation breast cancer progressing after a non-steroidal aromatase inhibitor.[8] To manage anticipated overlapping toxicities, a lower dose of ridaforolimus (10 mg vs 30 mg) was used in the triplet R/D/E arm.[15] The trial failed to meet its primary endpoint; median PFS was numerically shorter in the R/D/E arm (23.3 weeks) compared to the R/E arm (31.9 weeks).[15] ORR was also lower in the triplet arm (15% vs 25%).[15] While the lower ridaforolimus dose in the R/D/E arm resulted in less stomatitis and pneumonitis, the lack of efficacy improvement suggests the dose reduction may have compromised the potential benefit of the mTOR inhibitor component.[15] Hyperglycemia rates were similar between arms.[15] Earlier Phase I studies involving Dalotuzumab in breast cancer included NCT00759785 (details limited) [42] and NCT00730379 (combination with ridaforolimus).[11] A meta-analysis also suggested potential harmful effects of Dalotuzumab in breast cancer subgroups.[19]

3.3.4 Other Solid Tumors and Early Phase Studies

Dalotuzumab monotherapy was evaluated in a Phase I dose-escalation study (NCT00694356) in 80 patients with various advanced, chemotherapy-refractory solid tumors.[3] Doses up to 20 mg/kg weekly were tested without identifying a maximum tolerated dose (MTD).[3] The drug was generally well-tolerated, with Grade 1-3 hyperglycemia occurring in 19% of patients, typically responsive to metformin.[20] Pharmacokinetic analysis showed dose-proportional exposure and a long terminal half-life (≥95 hours at doses ≥5 mg/kg), supporting less frequent dosing intervals.[20] Pharmacodynamic assessments confirmed inhibition of IGF-1R pathway signaling in tumor biopsies, and some clinical activity (stable disease, partial responses) was observed.[20]

Combination strategies targeting multiple pathways were explored in Phase I (NCT01233168), combining Dalotuzumab with an AKT inhibitor (MK-2206), an mTOR inhibitor (ridaforolimus), or a Notch inhibitor (MK-0752).[39] These combinations were deemed tolerable, although dose-limiting toxicities (DLTs) including neutropenia, rash, gastrointestinal issues, and dehydration were encountered.[39] Efficacy appeared limited, with only seven patients remaining on study beyond four cycles, reinforcing the need for predictive biomarkers.[41]

In metastatic pancreatic cancer, a Phase I/II study combined Dalotuzumab with gemcitabine, with or without erlotinib.[16] The combinations were tolerable, with MTDs established (10 mg/kg with gemcitabine, 5 mg/kg with gemcitabine/erlotinib). Interestingly, the addition of Dalotuzumab to gemcitabine was associated with an improvement in OS compared to gemcitabine plus erlotinib, although PFS was not different. Tissue and serum IGF-1 levels did not correlate with outcomes in this study.[16]

3.4 Analysis of Combination Strategies

The clinical development program heavily relied on combination approaches, aiming to enhance efficacy by targeting complementary pathways or overcoming resistance mechanisms.

  • Chemotherapy Combinations: Combining Dalotuzumab with cytotoxic agents (pemetrexed/cisplatin in NSCLC [4], irinotecan in mCRC [6], etoposide/cisplatin in SCLC [17], gemcitabine in pancreatic cancer [16]) was based on potential synergistic effects observed preclinically.[6] While generally feasible from a safety perspective, these combinations consistently added hyperglycemia toxicity without providing a clear efficacy advantage over chemotherapy alone.[5]
  • EGFR Inhibitor Combinations: Targeting both IGF-1R and EGFR (with cetuximab in mCRC [6] or erlotinib in NSCLC [14]) was rationalized by known pathway cross-talk and potential co-dependence in tumors.[1] Despite the biological rationale, these combinations failed to improve clinical outcomes in randomized trials.[14]
  • PI3K/Akt/mTOR Pathway Inhibitor Combinations: Combining Dalotuzumab with mTOR inhibitors (ridaforolimus [8]) or an AKT inhibitor (MK-2206 [39]) aimed to counteract feedback activation of Akt signaling often seen with mTOR inhibition, a known resistance mechanism.[15] While biologically sound and tolerable in Phase I [39], the Phase II breast cancer trial combining Dalotuzumab with ridaforolimus/exemestane did not show benefit, potentially confounded by the need to use a lower, possibly sub-therapeutic, dose of ridaforolimus due to toxicity concerns.[15]
  • Notch Inhibitor Combination: The combination with the Notch inhibitor MK-0752 was based on preclinical evidence of cross-talk between IGF-1R and Notch signaling.[39] This combination was tolerable in Phase I but showed limited efficacy.[39]

The consistent failure of these varied combination strategies suggests that simply adding Dalotuzumab to existing therapies was insufficient to overcome the complex resistance mechanisms operating in unselected patient populations. The lack of significant efficacy improvements points towards the need for more sophisticated approaches, heavily reliant on predictive biomarkers to identify responsive patient subsets, rather than broad application of these combinations.

3.5 Overall Safety and Tolerability Profile

Across numerous trials, Dalotuzumab was generally considered well-tolerated as monotherapy and feasible in combination regimens.[2] However, a distinct pattern of adverse events (AEs) emerged:

  • Hyperglycemia: This was the most characteristic AE associated with Dalotuzumab, observed frequently across different trials and combinations.[1] It was typically Grade 1-3 in severity and often manageable with oral agents like metformin or occasionally insulin.[14] This AE serves as in vivo confirmation of target engagement and interference with the IGF/insulin signaling axis.
  • Common AEs: Other frequently reported AEs included asthenia/fatigue, nausea, diarrhea, skin toxicities (rash, dermatitis acneiform - often linked to EGFR inhibitors), decreased appetite, and stomatitis (prominent with ridaforolimus combination).[1] Hematologic toxicities were common when combined with chemotherapy.[4]
  • Serious AEs (SAEs): While less common, SAEs reported in various trials included febrile neutropenia, sepsis, thrombosis, dehydration, pneumonitis (with ridaforolimus), leukocytoclastic vasculitis (rare, resolved on discontinuation), and a serum sickness-like reaction (rare).[3] Some treatment-related deaths occurred in combination studies, often attributed to complications of chemotherapy or underlying disease progression.[4]

The consistent signal of hyperglycemia confirms on-target activity but also represents a significant clinical challenge. As discussed in relation to other IGF-1R inhibitors [26], interference with this pathway can exacerbate underlying insulin resistance or metabolic issues. This toxicity may have practical implications, potentially limiting the achievable dose intensity in combination regimens (as suggested in the ridaforolimus combination trial [15]) and complicating the management of patients, particularly those with pre-existing diabetes or metabolic syndrome.

4. Development Outcome and Analysis

4.1 Summary of Efficacy Findings

Despite extensive clinical investigation, Dalotuzumab failed to demonstrate clinically meaningful efficacy across its major target indications. Randomized Phase II/III trials in KRAS wild-type mCRC (NCT00614393) [18], refractory advanced NSCLC (NCT00654420) [14], first-line non-squamous NSCLC (NCT00799240) [22], and advanced ER+ breast cancer (NCT01605396) [15] consistently showed that adding Dalotuzumab to standard therapies did not significantly improve primary endpoints like PFS or OS. In some cases, particularly in the mCRC and NSCLC + erlotinib trials, there were numerical trends suggesting potentially worse OS in the Dalotuzumab-containing arms.[14] Objective response rates were generally not improved.[14] While early Phase I monotherapy studies reported some instances of stable disease or partial responses [20], and the Phase I SCLC combination trial showed a high ORR [17], these signals did not translate into success in larger, controlled trials. Meta-analysis data further supported the overall lack of benefit and even suggested potential harm in certain subgroups, such as breast and colorectal cancer.[19]

4.2 Evidence for Discontinuation of Development

Multiple lines of evidence strongly indicate that the clinical development of Dalotuzumab (MK-0646) was discontinued by Merck:

  • Trial Terminations for Futility: The pivotal Phase II/III trial in mCRC (NCT00614393) was prematurely stopped following an interim analysis because the Dalotuzumab arms failed to meet predefined criteria for continuation, indicating a low probability of success.[18] The Phase IIA trial in biomarker-selected metastatic rectal cancer (NCT01609231) was also terminated.[16]
  • Lack of Recent Development: No recent reports of active development have been identified for Dalotuzumab in indications like Crohn's disease or delayed graft function, where it had reached Phase I or II previously.
  • Absence from Sponsor Pipeline: Dalotuzumab (MK-0646) was listed as an asset in Merck's pipeline in regulatory filings from 2011 and 2012.[24] However, it is notably absent from more recent pipeline reports issued by GSK (which collaborates with Merck on some oncology assets but Dalotuzumab was a Merck program) and Merck's own clinical trial listings for relevant indications. While Wikipedia [1] mentions ongoing trials, this appears outdated given the published negative results and pipeline omissions.
  • Class-Wide Challenges: The discontinuation aligns with a broader trend observed for other anti-IGF-1R antibodies, such as figitumumab, which were also halted due to lack of efficacy or unacceptable toxicity in large trials.[26]

4.3 Analysis of Potential Reasons for Lack of Clinical Success

The discrepancy between the strong preclinical rationale for targeting IGF-1R and the disappointing clinical results of Dalotuzumab likely stems from several interconnected factors, reflecting the challenges inherent in targeting this complex pathway [26]:

  • Pathway Complexity and Redundancy: The IGF axis involves intricate cross-talk, particularly with the Insulin Receptor (IR) pathway. IGF-1R and IR can form hybrid receptors, and the IR itself (especially the fetal IR-A isoform) can bind IGF-2 and mediate growth signals.[26] Dalotuzumab's specificity for IGF-1R, while potentially reducing metabolic toxicity, likely allowed cancer cells to bypass the blockade via signaling through IR homodimers or IGF-1R/IR hybrids.[26] This inherent redundancy may render IGF-1R inhibition alone insufficient.
  • Lack of Predictive Biomarkers: A major hurdle was the inability to reliably identify patients most likely to benefit from Dalotuzumab. Initial hypotheses often centered on IGF-1R expression levels, but IHC assessment proved inconsistent and poorly predictive of response.[3] Efforts to explore other biomarkers, such as circulating ligands (IGF-1, IGF-2) or downstream pathway markers, yielded intriguing but ultimately inconclusive or unvalidated results.[16] Without robust biomarkers, trials enrolled unselected populations where potential benefit in a small subset could be masked by lack of efficacy in the majority.
  • Endocrine Disruption and On-Target Toxicity: IGF-1R plays a role in the negative feedback regulation of growth hormone (GH) secretion. Blocking IGF-1R can disrupt this feedback, leading to increased GH and consequently elevated circulating IGF-1 levels.[26] This compensatory increase could potentially activate remaining unblocked receptors (IR, hybrids) or even contribute to resistance. Furthermore, the resulting endocrine disruption can exacerbate insulin resistance, leading to the frequently observed hyperglycemia AE.[1] This toxicity, while often manageable, could complicate treatment, particularly in patients with pre-existing metabolic conditions, and may have limited the ability to combine Dalotuzumab effectively at optimal doses with other agents.[15]
  • Resistance Mechanisms: Beyond pathway redundancy, cancer cells can develop resistance through various mechanisms, including activation of alternative growth signaling pathways (e.g., PI3K/Akt/mTOR activation independent of IGF-1R, or Notch signaling).[38] The feedback activation of AKT observed upon mTOR inhibition, mediated via sustained IGF1R/IRS1 signaling, exemplifies how targeting one node can trigger compensatory mechanisms that limit efficacy, a challenge the Dalotuzumab/ridaforolimus combination failed to overcome clinically.[15]
  • Suboptimal Combination Strategies: While combining Dalotuzumab with chemotherapy or other targeted agents was biologically rationalized, these strategies failed to yield clinical benefit. This could be due to an inability to achieve sufficient synergistic pathway inhibition at tolerable doses, the selection of inappropriate combination partners for the specific tumor biology, or the lack of patient stratification based on combination-specific biomarkers.[14]

The failure of Dalotuzumab and its contemporaries underscores that potent target engagement and a strong preclinical rationale are insufficient for clinical success when targeting complex, physiologically vital pathways like IGF signaling. The inability to overcome pathway redundancy and feedback mechanisms, coupled with the lack of predictive biomarkers for patient selection, proved insurmountable obstacles.

4.4 Biomarker Exploration

Recognizing the likely need for patient selection, several biomarker strategies were explored, albeit with limited success:

  • IGF-1R Expression: Tumor expression of IGF-1R, typically assessed by immunohistochemistry (IHC), was investigated in early trials [3] but did not emerge as a reliable predictive marker.[26]
  • Ligand Levels (IGF-1, IGF-2): Circulating or tumor mRNA levels of IGF ligands were explored. In the Phase II/III mCRC trial (NCT00614393), high tumor IGF-1 mRNA expression showed a complex association: it numerically correlated with better PFS/OS in patients receiving Dalotuzumab (Arm A) compared to placebo (Arm C), but also predicted poorer outcomes (lower response, shorter PFS) in the placebo/control arm (receiving cetuximab/irinotecan).[18] This suggests IGF-1 levels might influence sensitivity to both IGF-1R and EGFR inhibition. Conversely, in a pancreatic cancer trial, IGF-1 levels did not correlate with outcome.[16] Low IGF-2 expression was used as an entry criterion for a terminated rectal cancer trial (NCT01609231) and explored in expansion cohorts of a Phase I combination study (NCT01233168).[39]
  • Downstream Pathway Markers: Phosphorylation status of downstream effectors like pMAPK and pAKT were assessed in Phase I pharmacodynamic studies, confirming pathway inhibition.[13] However, their utility as predictive markers in larger trials was not established.
  • Other Signatures: A "low-RAS signature" was used to select ovarian cancer patients for an expansion cohort in the Phase I combination trial NCT01233168.[39]

The exploratory finding in mCRC regarding IGF-1 mRNA levels [18] is particularly noteworthy. It suggests that the status of the IGF pathway might not only predict response to IGF-1R inhibitors but could also modulate sensitivity to other targeted therapies like EGFR inhibitors. This highlights a potential role for IGF pathway biomarkers in optimizing existing treatment strategies, even beyond the context of direct IGF-1R targeting. However, overall, the biomarker efforts for Dalotuzumab did not yield a validated strategy for prospective patient selection.

5. Conclusion

Dalotuzumab (MK-0646) represents a rationally designed, humanized monoclonal antibody targeting the IGF-1R, developed based on compelling preclinical evidence implicating the IGF pathway in cancer pathogenesis.[1] Its clinical development program was extensive, evaluating the drug across multiple tumor types and in various combination regimens.[8]

Despite demonstrating target engagement, evidenced by pharmacodynamic effects and the characteristic adverse event of hyperglycemia [14], Dalotuzumab consistently failed to improve primary efficacy endpoints such as PFS and OS in key randomized Phase II and III trials when added to standard-of-care therapies in mCRC, NSCLC, and breast cancer.[14] The combination regimens were generally feasible but offered no significant clinical advantage over standard therapy alone.

Based on the reported trial outcomes, including premature termination for futility [18], and its absence from recent sponsor pipeline disclosures, the clinical development of Dalotuzumab appears to have been discontinued.

The experience with Dalotuzumab provides valuable lessons for oncology drug development, particularly when targeting complex signaling pathways with crucial physiological roles. It underscores that:

  1. Pathway complexity and redundancy (e.g., IGF-1R vs. IR signaling, feedback loops) can significantly limit the efficacy of highly specific inhibitors.[26]
  2. Robust, validated predictive biomarkers are essential for identifying patient populations likely to benefit from targeted therapies, especially when efficacy signals are modest or absent in unselected populations.[18]
  3. On-target toxicities (like hyperglycemia with IGF-1R inhibition) must be anticipated and managed, as they can impact tolerability, limit dosing in combinations, and potentially affect patient outcomes.[15]

In conclusion, Dalotuzumab serves as a well-documented example of an investigational targeted cancer therapy that, despite a strong scientific rationale and successful target engagement, failed to translate preclinical promise into demonstrable clinical benefit. Its development journey highlights the significant challenges in targeting the IGF pathway and contributes important, albeit negative, data to the field, emphasizing the critical need for deeper biological understanding and effective patient stratification strategies in modern oncology.

Works cited

  1. Dalotuzumab - Wikipedia, accessed May 2, 2025, https://en.wikipedia.org/wiki/Dalotuzumab
  2. (PDF) Dalotuzumab, a recombinant humanized mAb targeted against IGFR1 for the treatment of cancer - ResearchGate, accessed May 2, 2025, https://www.researchgate.net/publication/44647327_Dalotuzumab_a_recombinant_humanized_mAb_targeted_against_IGFR1_for_the_treatment_of_cancer
  3. Inhibition of insulin-like growth factor receptor: end of a targeted therapy? - Pillai, accessed May 2, 2025, https://tlcr.amegroups.org/article/view/652/1405
  4. Impact Study: MK-0646 (Dalotuzumab), Insulin Growth Factor 1 Receptor Antibody Combined with Pemetrexed and Cisplatin in Stage IV Metastatic Non-squamous Lung Cancer, accessed May 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4710681/
  5. MK-0646 (Dalotuzumab), Insulin Growth Factor 1 Receptor Antibody Combined with Pemetrexed and Cisplatin in Stage I - CORE, accessed May 2, 2025, https://core.ac.uk/download/pdf/82888987.pdf
  6. Phase 1 pharmacokinetic study of MK-0646 (dalotuzumab), an anti-insulin-like growth factor-1 receptor monoclonal antibody, in combination with cetuximab and irinotecan in Japanese patients with advanced colorectal cancer - PubMed Central, accessed May 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3753462/
  7. Dalotuzumab: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed May 2, 2025, https://go.drugbank.com/drugs/DB11840
  8. Dalotuzumab Completed Phase 2 Trials for Breast Neoplasms Treatment | DrugBank Online, accessed May 2, 2025, https://go.drugbank.com/drugs/DB11840/clinical_trials?conditions=DBCOND0028011&phase=2&purpose=treatment&status=completed
  9. Dalotuzumab Completed Phase 2 Trials for Non-Small Cell Lung Carcinoma Treatment, accessed May 2, 2025, https://go.drugbank.com/drugs/DB11840/clinical_trials?conditions=DBCOND0034623&phase=2&purpose=treatment&status=completed
  10. dalotuzumab - DrugMapper, accessed May 2, 2025, https://drugmapper.helsinki.fi/CompoundInfo?compound_id=2126383
  11. Neoplastic Syndrome Completed Phase 1 Trials for Dalotuzumab (DB11840) - DrugBank, accessed May 2, 2025, https://go.drugbank.com/indications/DBCOND0028594/clinical_trials/DB11840?phase=1&status=completed
  12. Colorectal Cancers Completed Phase 1 Trials for Dalotuzumab (DB11840) - DrugBank, accessed May 2, 2025, https://go.drugbank.com/indications/DBCOND0063246/clinical_trials/DB11840?phase=1&status=completed
  13. A Randomized Phase II/III Study of Dalotuzumab in Combination With Cetuximab and Irinotecan in Chemorefractory, KRAS Wild-Type, - Oxford Academic, accessed May 2, 2025, https://academic.oup.com/jnci/article-pdf/doi/10.1093/jnci/djv258/17985755/djv258.pdf
  14. Activity of dalotuzumab, a selective anti-IGF1R antibody, in ..., accessed May 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4237770/
  15. A randomized phase II trial of ridaforolimus, dalotuzumab, and exemestane compared with ridaforolimus and exemestane in patients with advanced breast cancer - PubMed Central, accessed May 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5709225/
  16. Dalotuzumab - Drug Targets, Indications, Patents - Patsnap Synapse, accessed May 2, 2025, https://synapse.patsnap.com/drug/132ba8c708ad4f0aa0f9a4cab175d213
  17. NCIC CTG IND.190 phase I trial of dalotuzumab (MK-0646) in combination with cisplatin and etoposide in extensive-stage small-cell lung cancer - PubMed, accessed May 2, 2025, https://pubmed.ncbi.nlm.nih.gov/24518092/
  18. Randomized Phase II/III Study of Dalotuzumab in Combination With ..., accessed May 2, 2025, https://academic.oup.com/jnci/article/107/12/djv258/2457719
  19. Update of IGF-1 receptor inhibitor (ganitumab, dalotuzumab, cixutumumab, teprotumumab and figitumumab) effects on cancer therapy - PMC, accessed May 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5438747/
  20. A Phase I, Pharmacokinetic and Pharmacodynamic Study of Dalotuzumab (MK-0646), an Anti-IGF-1R Monoclonal Antibody, in Patients with Advanced Solid Tumors - ResearchGate, accessed May 2, 2025, https://www.researchgate.net/publication/51541907_A_Phase_I_Pharmacokinetic_and_Pharmacodynamic_Study_of_Dalotuzumab_MK-0646_an_Anti-IGF-1R_Monoclonal_Antibody_in_Patients_with_Advanced_Solid_Tumors
  21. A phase I pharmacokinetic and pharmacodynamic study of dalotuzumab (MK-0646), an anti-insulin-like growth factor-1 receptor monoclonal antibody, in patients with advanced solid tumors - PubMed, accessed May 2, 2025, https://pubmed.ncbi.nlm.nih.gov/21810918/
  22. Impact Study: MK-0646 (Dalotuzumab), Insulin Growth Factor 1 Receptor Antibody Combined with Pemetrexed and Cisplatin in Stage IV Metastatic Non-squamous Lung Cancer - Frontiers, accessed May 2, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2015.00301/full
  23. Impact Study: MK-0646 (Dalotuzumab), Insulin Growth Factor 1 Receptor Antibody Combined with Pemetrexed and Cisplatin in Stage IV Metastatic Non-squamous Lung Cancer - PubMed, accessed May 2, 2025, https://pubmed.ncbi.nlm.nih.gov/26793618/
  24. Form 10-Q - SEC.gov, accessed May 2, 2025, https://www.sec.gov/Archives/edgar/data/310158/000119312512463586/d400394d10q.htm
  25. e10vk - SEC.gov, accessed May 2, 2025, https://www.sec.gov/Archives/edgar/data/310158/000095012311019286/y87773e10vk.htm
  26. Insulin-like Growth Factor Receptor Inhibitors: Baby or the Bathwater ..., accessed May 2, 2025, https://academic.oup.com/jnci/article/104/13/975/2516935
  27. Targeting the Insulin Growth Factor Pathway in Gastrointestinal Cancers, accessed May 2, 2025, https://www.cancernetwork.com/view/targeting-insulin-growth-factor-pathway-gastrointestinal-cancers
  28. DALOTUZUMAB - Inxight Drugs, accessed May 2, 2025, https://drugs.ncats.io/drug/6YI1L648RH
  29. Dalotuzumab (Anti-IGF1R / CD221) CD markers antibody | Read Reviews & Product Use Citations - Selleckchem.com, accessed May 2, 2025, https://www.selleckchem.com/products/dalotuzumab-anti-igf1r-cd221.html
  30. https://rxnav.nlm.nih.gov/REST/rxcui/1432293/allProperties.xml?prop\=all, accessed May 2, 2025, https://rxnav.nlm.nih.gov/REST/rxcui/1432293/allProperties.xml?prop=all
  31. Dalotuzumab Biosimilar(Anti-IGF1R / CD221 Reference Antibody) - Biorbyt, accessed May 2, 2025, https://www.biorbyt.com/index.php/dalotuzumab-biosimilaranti-ig-orb2703822.html
  32. Dalotuzumab - Biorbyt, accessed May 2, 2025, https://www.biorbyt.com/index.php/dalotuzumab-orb1980643.html
  33. Buy research grade Dalotuzumab biosimilar ,anti-MK-0646 antibody - AntibodySystem, accessed May 2, 2025, https://www.antibodysystem.com/product/10044.html
  34. Dalotuzumab Biosimilar – Anti-IGF1R, CD221 mAb – Research Grade - ProteoGenix, accessed May 2, 2025, https://www.proteogenix.science/product/dalotuzumab-biosimilar-anti-igf1r-cd221-mab-research-grade/
  35. Dalotuzumab | CAS NO.:1005389-60-5 | GlpBio, accessed May 2, 2025, https://www.glpbio.com/sp/dalotuzumab.html
  36. Dalotuzumab (MK-0646) | Humanized Anti-IGF1R mAb - MedchemExpress.com, accessed May 2, 2025, https://www.medchemexpress.com/dalotuzumab.html
  37. Dalotuzumab, a recombinant humanized mAb targeted against IGFR1 for the treatment of cancer - PubMed, accessed May 2, 2025, https://pubmed.ncbi.nlm.nih.gov/20521225/
  38. IGF-1R and mTOR Blockade: Novel Resistance Mechanisms and Synergistic Drug Combinations for Ewing Sarcoma - PMC - PubMed Central, accessed May 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5241894/
  39. A parallel-arm phase I trial of the humanised anti-IGF-1R antibody dalotuzumab in combination with the AKT inhibitor MK-2206, the mTOR inhibitor ridaforolimus, or the NOTCH inhibitor MK-0752, in patients with advanced solid tumours - PubMed Central, accessed May 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4229637/
  40. Combination of the mTOR Inhibitor Ridaforolimus and the Anti-IGF1R Monoclonal Antibody Dalotuzumab: Preclinical Characterization and Phase I Clinical Trial - PubMed Central, accessed May 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5705197/
  41. A parallel-arm phase I trial of the humanised anti-IGF-1R antibody dalotuzumab in combination with the AKT inhibitor MK-2206, the mTOR inhibitor ridaforolimus, or the NOTCH inhibitor MK-0752, in patients with advanced solid tumours - PubMed, accessed May 2, 2025, https://pubmed.ncbi.nlm.nih.gov/25290091/
  42. A Study of Dalotuzumab (MK-0646) in Breast Cancer Patients (MK, accessed May 2, 2025, https://clin.larvol.com/trial/NCT00759785
  43. Dalotuzumab Completed Phase 1 Trials for Non-Small Cell Lung Carcinoma Treatment, accessed May 2, 2025, https://go.drugbank.com/drugs/DB11840/clinical_trials?conditions=DBCOND0030527&phase=1&purpose=treatment&status=completed
  44. A Randomized Phase II/III Study of Dalotuzumab in Combination With Cetuximab and Irinotecan in Chemorefractory, KRAS Wild-Type, Metastatic Colorectal Cancer - PubMed, accessed May 2, 2025, https://pubmed.ncbi.nlm.nih.gov/26405092/
  45. A randomized phase II/III study of the anti-IGF-1R antibody MK-0646 (dalotuzumab) in combination with cetuximab (Cx) and irinotecan (Ir) in the treatment of chemorefractory metastatic colorectal cancer (mCRC) with wild-type (wt) KRAS status. - ResearchGate, accessed May 2, 2025, https://www.researchgate.net/publication/313942083_A_randomized_phase_IIIII_study_of_the_anti-IGF-1R_antibody_MK-0646_dalotuzumab_in_combination_with_cetuximab_Cx_and_irinotecan_Ir_in_the_treatment_of_chemorefractory_metastatic_colorectal_cancer_mCRC_
  46. Study of Dalotuzumab (MK-0646) in Combination With Cetuximab and Irinotecan in Metastatic Colorectal Cancer (MK-0646-004) - LarvolClin, accessed May 2, 2025, https://clin.larvol.com/trial-detail/NCT00614393
  47. Phase I Study of Dalotuzumab (MK-0646) in Combination With Cetuximab and Irinotecan in Participants With Colorectal Cancer (MK-0646-016) - LarvolClin, accessed May 2, 2025, https://clin.larvol.com/trial-detail/NCT00925015
  48. EU Clinical Trials Register, accessed May 2, 2025, https://www.clinicaltrialsregister.eu/ctr-search/trial/2012-000317-36/DK
  49. Merck, “A Study Evaluating Dalotuzumab (MK0646) in Combination, accessed May 2, 2025, https://www.scirp.org/(S(ny23rubfvg45z345vbrepxrl))/reference/referencespapers?referenceid=662176
  50. Dalotuzumab in Metastatic Non-squamous Lung Cancer - Frontiers, accessed May 2, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2015.00301/epub
  51. A Combination Study With Ridaforolimus (MK8669 ... - ScanMedicine, accessed May 2, 2025, https://scanmedicine.com/clinicaltrials/NCT00730379
  52. Combination of the mTOR Inhibitor Ridaforolimus and the Anti-IGF1R Monoclonal Antibody Dalotuzumab: Preclinical Characterization and Phase I Clinical Trial - AACR Journals, accessed May 2, 2025, https://aacrjournals.org/clincancerres/article/21/1/49/13917/Combination-of-the-mTOR-Inhibitor-Ridaforolimus

Published at: May 2, 2025

This report is continuously updated as new research emerges.

© Copyright 2025. All Rights Reserved by MedPath