997 results on '"*MAYTANSINE"'
Search Results
152. A Study of Trastuzumab Emtansine (T-DM1) Sequentially With Anthracycline-based Chemotherapy, as Adjuvant or Neoadjuvant Therapy for Patients With Early Stage Herceptin (HER)2-positive Breast Cancer
- Published
- 2014
153. Biological evaluation of 9-thioansamitocin P3.
- Author
-
Vasilevich, Natalya I., Jiang, Huangyu, Xiao, Haihua, Feng, Kunxian, Jian, Chengfang, Chen, Changfeng, Li, Min, Chen, Zhenhua, Pang, Li, Li, Xiang, Chestkov, Alexander V., Sun, Andre H., Xu, Wang, Fuselier, Joseph A., Coy, David H., and Sun, Lichun
- Subjects
- *
TUMOR growth , *DRUG delivery systems , *LIVER microsomes , *AMES test , *MUTAGENICITY testing - Abstract
Highly cytotoxic maytansine derivatives are widely used in targeted tumor delivery. Structure-activity studies published earlier suggested the C9 carbinol to be a key element necessary to retain the potency. However, in 1984 a patent was published by Takeda in which the synthesis of 9-thioansamitocyn (AP3SH) was described and its activity in xenograft models was shown. In this article we summarize the results of an extended study of the anti-tumor properties of AP3SH. Like other maytansinoids, it induces apoptosis and arrests the cell cycle in the G2/M phase. It is metabolized in liver microsomes predominately by C3A4 isoform and doesn't inhibit any CYP isoforms except CYP3A4 (midazolam, IC50 7.84 μM). No hERG inhibition, CYP induction or mutagenicity in Ames tests were observed. AP3SH demonstrates high antiproliferative activity against 25 tumor cell lines and tumor growth inhibition in U937 xenograft model. Application of AP3SH as a cytotoxic payload in drug delivery system was demonstrated by us earlier. • 9-Thioansamitocin-P3 (AP3SH) was synthesized and its anti-tumor activity was studied. • AP3SH shows anti-tumor activity in a panel of in vitro assays. • AP3SH suppresses tumor growth in U937 xenograft mice model. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
154. A Study of Trastuzumab Emtansine (Trastuzumab-MCC-DM1, T-DM1) in Combination With Pertuzumab Administered to Patients With Human Epidermal Growth Factor Receptor-2 (HER2)-Positive Locally Advanced or Metastatic Breast Cancer Who Have Previously Received Trastuzumab
- Author
-
Roche Pharma AG
- Published
- 2013
155. A Study of the Efficacy and Safety of Trastuzumab Emtansine (Trastuzumab-MCC-DM1) vs. Trastuzumab (Herceptin®) and Docetaxel (Taxotere®) in Patients With Metastatic HER2-positive Breast Cancer Who Have Not Received Prior Chemotherapy for Metastatic Disease
- Published
- 2013
156. Corrected QT Interval Effects of Trastuzumab Emtansine (T-DM1) in Patients With Human Epidermal Growth Factor Receptor 2 (HER2)-Positive Locally Advanced or Metastatic Breast Cancer and the Safety and Tolerability of Combined T-DM1 and Pertuzumab in Patients With Early Disease Progression
- Published
- 2013
157. A Study of Trastuzumab Emtansine (Trastuzumab-MCC-DM1) Administered Intravenously to Patients With Human Epidermal Growth Factor Receptor 2 (HER2)-Positive Metastatic Breast Cancer
- Published
- 2013
158. A Review on Phytochemicals of the Genus Maytenus and Their Bioactive Studies
- Author
-
Yuan-Yuan Huang, Lu Chen, Guo-Xu Ma, Xu-Dong Xu, Xue-Gong Jia, Fu-Sheng Deng, Xue-Jian Li, and Jing-Quan Yuan
- Subjects
Maytenus ,triterpenoids ,sesquiterpenes ,alkaloids ,synthesis of maytansine ,Organic chemistry ,QD241-441 - Abstract
The genus Maytenus is a member of the Celastraceae family, of which several species have long been used in traditional medicine. Between 1976 and 2021, nearly 270 new compounds have been isolated and elucidated from the genus Maytenus. Among these, maytansine and its homologues are extremely rare in nature. Owing to its unique skeleton and remarkable bioactivities, maytansine has attracted many synthetic endeavors in order to construct its core structure. In this paper, the current status of the past 45 years of research on Maytenus, with respect to its chemical and biological activities are discussed. The chemical research includes its structural classification into triterpenoids, sesquiterpenes and alkaloids, along with several chemical synthesis methods of maytansine or maytansine fragments. The biological activity research includes activities, such as anti-tumor, anti-bacterial and anti-inflammatory activities, as well as HIV inhibition, which can provide a theoretical basis for the better development and utilization of the Maytenus.
- Published
- 2021
- Full Text
- View/download PDF
159. Maytansine-bearing antibody-drug conjugates induce in vitro hallmarks of immunogenic cell death selectively in antigen-positive target cells
- Author
-
Maxine Bauzon, Penelope M. Drake, Robyn M. Barfield, Brandon M. Cornali, Igor Rupniewski, and David Rabuka
- Subjects
immunogenic cell death ,antibody-drug conjugate ,adc ,maytansine ,immunooncology ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Oncology treatment has been revolutionized by the introduction of immune checkpoint inhibitor drugs, which enable 20–40% of patients to generate anti-tumor immune responses. Combination treatment approaches with chemotherapeutic drugs may enable responses in the remaining patient cohorts. In this regard, a handful of drugs are promising due to their ability to induce immunogenic cell death in target cells. However, these agents are systemically delivered and indiscriminately cytotoxic to proliferating cells. By contrast, antibody-drug conjugates can selectively deliver a cytotoxic payload to a tumor, sparing most healthy cells. The ability of antibody-drug conjugates to induce immunogenic cell death in target cells has not yet been determined, although preclinical in vivo studies suggest this possibility. Here, we describe for the first time production of the in vitro hallmarks of immunogenic cell death – ecto-calreticulin and secreted ATP and HMGB1 protein – by cells in response to treatment with antibody-drug conjugates bearing a maytansine payload.
- Published
- 2019
- Full Text
- View/download PDF
160. Development and validation of pharmacokinetics assays for a novel HER2-targeting antibody-drug conjugate (SHR-A1201): Application to its dose-escalation pharmacokinetic study.
- Author
-
Li X, Wang Y, Hu W, Song Q, and Ding L
- Subjects
- Humans, Female, Ado-Trastuzumab Emtansine, Chromatography, Liquid, Antibodies, Monoclonal, Humanized pharmacokinetics, Receptor, ErbB-2 metabolism, Tandem Mass Spectrometry, Trastuzumab therapeutic use, Cytotoxins, Immunoconjugates therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Maytansine
- Abstract
Approximately 25% of breast cancer patients with HER2 overexpression tend to have a high risk of disease progression and death. Various HER2-targeting therapies have been approved for treatment. Recently, a novel antibody-drug conjugate, SHR-A1201, is being researched and developed. For the pharmacokinetic study of SHR-A1201, suitable bioanalytical methods are needed for quantifying unconjugated cytotoxin, cytotoxin-conjugated antibodies and total antibodies. In this research, bioanalytical methods involving a highly sensitive LC-MS/MS assay for unconjugated cytotoxic payload DM1 in human plasma, ELISA strategies for DM1-conjugated trastuzumab and total trastuzumab in human serum were developed, validated and successfully applied to a phase I dose-escalation pharmacokinetic study of SHR-A1201. The pharmacokinetic properties and exposure-to-dose proportionality was evaluated for SHR-A1201. According to the bioanalytical method validation guidance, the bioanalytical methods were fully validated and the validation results met the acceptance criteria. The nonspecific binding of DM1 and dimer was avoided for the LC-MS/MS assay. In the dose-escalation pharmacokinetic study of SHR-A1201, a potential dose-proportional pharmacokinetics was observed over the dose from 1.2 mg/kg to 4.8 mg/kg. The validated bioanalytical strategies are robust and reproducible and these bioanalytical methods will contribute to better understanding of the pharmacokinetic properties of SHR-A1201., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
161. Plant Endophytes and Epiphytes: Burgeoning Sources of Known and "Unknown" Cytotoxic and Antibiotic Agents?
- Author
-
Newman, David J. and Cragg, Gordon M.
- Subjects
- *
ANTIBIOTICS , *ANTINEOPLASTIC agents , *PLANT chemical analysis , *ALKALOIDS , *CAMPTOTHECIN , *FUNGI , *LEAVES , *MACROLIDE antibiotics , *MOLECULAR structure , *MYCOTOXINS , *PACLITAXEL , *BENZENE derivatives , *PHARMACODYNAMICS - Abstract
In the last 20 or so years, the influence of endophytes and, quite recently, epiphytes of plants upon the compounds found in those plants, which were usually assumed to be phytochemicals produced by the plant for a variety of reasons, often as a defense against predators, is becoming more evident, in particular in the case of antitumor agents originally isolated from plant sources, though antibiotic agents might also be found, particularly from epiphytes. In this review, we started with the first report in 1993 of a taxol-producing endophyte and then expanded the compounds discussed to include camptothecin, the vinca alkaloids, podophyllotoxin, and homoharringtonine from endophytic microbes and then the realization that maytansine is not a plant secondary metabolite at all, and that even such a well-studied plant such as Arabidopsis thaliana has a vast repertoire of potential bioactive agents in its leaf epiphytic bacteria. We have taken data from a variety of sources, including a reasonable history of these discoveries that were not given in recent papers by us, nor in other papers covering this topic. The sources included the Scopus database, but we also performed other searches using bibliographic tools, thus, the majority of the papers referenced are the originals, though we note some very recent papers that have built on previous results. We concluded with a discussion of the more modern techniques that can be utilized to "persuade" endophytes and epiphytes to switch on silent biosynthetic pathways and how current analytical techniques may aid in evaluating such programs. We also comment at times on some findings, particularly in the case of homoharringtonine, where there are repetitious data reports differing by a few years claiming the same endophyte as the producer. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
162. Antibody-drug conjugates of 7-ethyl-10-hydroxycamptothecin: Sacituzumab govitecan and labetuzumab govitecan.
- Author
-
Dong, Wenjuan, Shi, Jianyou, Yuan, Ting, Qi, Baowen, Yu, Jiying, Dai, Jingying, and He, Lin
- Subjects
- *
ANTIBODY-enzyme conjugates , *CYTOTOXINS , *STATINS (Cardiovascular agents) , *MAYTANSINE , *TUBULINS - Abstract
Abstract Monoclonal antibody (mAb), cytotoxins, and linker technology are three essential elements for developing a successful antibody-drug conjugate (ADC). In the research and development of ADCs industry, selected cytotoxins, such as auristatins and maytansines, are commonly tubulin inhibitors which are widely put into clinical use. Thereafter, with the booming development of ADCs, a large number of pharmaceutical companies have expanded a wide range of selectable cytotoxin product lines as well. Recently, the cytotoxic substance of 7-ethyl-10-hydroxycamptothecin (SN-38) conjugated to the monoclonal antibody by linker technology is developed as the second-generation ADCs. Here, the SN-38 families together with sacituzumab govitecan and labetuzumab govitecan are reviewed, whose features of metabolic pathway and toxicity in vivo are well-known. In sum, these methodology and technology would be convenient and flexible to be applied for developing the novel class of cytotoxins in ADCs industry. Graphical abstract Image 1 Highlights • The antibody-drug conjugates accurately target the tumors, carrying cytotoxins to eliminate the tumor cells. • Moderately-potent SN-38 as payloads have increased the drug-antibody ratio and the therapeutic index. • Sacituzumab govitecan can be used for treatment of various solid tumors, especially triple negative breast cancer. • Labetuzumab govitecan shows significantly higher anti-cancer effects for colorectal cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
163. Revival of a potent therapeutic maytansinoid agent using a strategy that combines covalent drug conjugation with sequential nanoparticle assembly.
- Author
-
Xie, Ke, Song, Shanshan, Zhou, Liqian, Wan, Jianqin, Qiao, Yiting, Wang, Min, Xie, Haiyang, Zhou, Lin, Zheng, Shusen, and Wang, Hangxiang
- Subjects
- *
NANOPARTICLES , *MAYTANSINE , *DRUG toxicity , *POLYETHYLENE glycol , *MOLECULAR weights , *COPOLYMERS - Abstract
Graphical abstract Abstract Maytansine and its related analogues are a class of highly potent anti-proliferation agents that have failed to be exploited as clinical drugs for human therapy due to unacceptable systemic toxicity. Here, we delineate a novel strategy that combines rational drug conjugation with subsequent nanoparticle assembly to systemically deliver this highly potent and toxic drug. To demonstrate this concept, we covalently coupled the thiolated maytansine derivative, the DM1 agent, to amphiphilic block co-polymers, polyethylene glycol (PEG)- block -polylactide (PLA), in varying molecular weights to generate two prodrug constructs (i.e., PEG 2K -PLA 2K -DM1 and PEG 2K -PLA 4K -DM1) via the maleimide-thiol reaction. The resulting two constructs are amenable to self-assembly in aqueous solutions and are systemically injectable for preclinical studies. In vivo evaluations indicate that PEG-PLA-DM1 conjugate-assembled nanoparticles (NPs) display substantially reduced drug toxicity compared to the free drug forms and NPs that physically encapsulate DM1. Furthermore, following systemic administration, these nanodrugs produced superior therapeutic efficacy over free DM1 in a colon tumor xenograft-bearing mouse model. Therefore, this study provides evidence that the conjugation of toxic drugs to assembling copolymers enables the alleviation of cancer drug toxicity and effective delivery of anticancer drugs. Thus, this DM1-formulated platform represents a new generation of nanotherapeutics that are available for further clinical evaluation. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
164. The Dawn of the Antibody–Drug Conjugates Era: How T-DM1 Reinvented the Future of Chemotherapy for Solid Tumors
- Author
-
Paolo Tarantino and Sara M. Tolaney
- Subjects
Cancer Research ,Immunoconjugates ,Oncology ,Humans ,Antineoplastic Agents ,Breast Neoplasms ,Female ,Maytansine ,Sulfides ,Trastuzumab ,Ado-Trastuzumab Emtansine ,Antibodies, Monoclonal, Humanized - Abstract
Delivering targeted chemotherapy through antibody–drug conjugates (ADC) has emerged as an extremely effective therapeutic strategy for multiple types of cancer. The first agent of this class to be established for treating a solid tumor was trastuzumab emtansine (T-DM1), approved in 2013 for the treatment of HER2-positive metastatic breast cancer. Much of the knowledge that led to this approval came from the landmark Cancer Research publication by Lewis Phillips and colleagues in 2008, where they described the in vitro and in vivo efficacy, pharmacokinetics, and toxicity of T-DM1, demonstrating its relevant preclinical activity against HER2-positive breast cancer models. In this article, the authors also explored the use of different linkers to conjugate the cytotoxic payload to the trastuzumab vehicle, demonstrating improved stability, efficacy, and tolerability of the compound when adopting a specific thioether linker. The findings from this work not only set the stage for the clinical development of T-DM1, but also highlighted the modularity of ADCs, with small changes in their components able to dramatically impact their activity and toxicity. This finding would prove key for the development of novel ADCs, several of which are now reshaping the way we treat breast cancer and other cancer types. In this commentary, we discuss the key implications of the work by Phillips and colleagues, putting it in context of the current and anticipated expansion in the use of ADCs to treat cancer. See related article by Phillips et al., Cancer Res 2008;68:9280–90.
- Published
- 2022
- Full Text
- View/download PDF
165. Comparison of the Efficacy, Safety, Pharmacokinetic and Immunogenicity of UJVIRA (ZRC-3256, Trastuzumab Emtansine) With the Kadcyla (Trastuzumab Emtansine) in the Treatment of HER2-Positive Metastatic Breast Cancer: A Randomized, Open-Label, Multicenter Study in India
- Author
-
Deven Parmar, Nirmal Raut, Rajnish Nagarkar, Ullas Batra, Tanveer Maksud, CT Satheesh, and Sanjeev Kumar
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Receptor, ErbB-2 ,India ,Breast Neoplasms ,Ado-Trastuzumab Emtansine ,Antibodies, Monoclonal, Humanized ,chemistry.chemical_compound ,Breast cancer ,Pharmacokinetics ,Internal medicine ,medicine ,Humans ,Maytansine ,business.industry ,Immunogenicity ,Absolute risk reduction ,Biosimilar ,Trastuzumab ,medicine.disease ,Metastatic breast cancer ,chemistry ,Trastuzumab emtansine ,Toxicity ,Female ,business - Abstract
Background: UJVIRA is the first DCGI approved biosimilar of trastuzumab emtansine (Kadcyla) which may offer an alternative cost-effective treatment option for HER2-positive metastatic breast cancer patients in India. This article summarizes the available clinical evidence supporting the biosimilarity of UJVIRA and Kadcyla with respect to efficacy, pharmacokinetic, safety, and immunogenicity. Methods: A phase 3, randomized, open-label, active-controlled study was conducted at 31 sites across India. A total of 168 patients were enrolled and randomized to receive either UJVIRA or Kadcyla. Of which, only first 50 patients were included in pharmacokinetic assessment. UJVIRA or Kadcyla were administered at a dose of 3.6 mg/kg by intravenous infusion every 3 weeks (21 days) for 8 cycles or until disease progression or unmanageable toxicity, whichever was earlier. The study assessed efficacy (ORR), safety, pharmacokinetics, and immunogenicity. Results: The ORR at the end of Week 24 was 37.76% in the UJVIRA and 33.33% in the Kadcyla group. The risk difference was 4.42% [-12.01, 20.85]. It met non-inferiority margin of -15%. The pharmacokinetic parameters were comparable between groups. No anti-drug antibody was detected in any of the treatment groups. The overall safety profile in terms of TEAEs and laboratory abnormalities was also comparable between the treatment groups. Conclusion: Results demonstrated biosimilarity between UJVIRA and Kadcyla in terms of efficacy, safety, pharmacokinetics, and immunogenicity. Therefore, UJVIRA could prove to be a cost-effective treatment alternative for HER2-positive metastatic breast cancer patients in India.
- Published
- 2022
- Full Text
- View/download PDF
166. Incidence of adverse events with therapies targeting HER2-positive metastatic breast cancer: a literature review
- Author
-
Edith A. Perez, Chau Dang, Caleb Lee, Jasmeet Singh, Kongming Wang, J. Bradley Layton, Alicia Gilsenan, Michelle D. Hackshaw, and Javier Cortes
- Subjects
Cancer Research ,Receptor, ErbB-2 ,Incidence ,Breast Neoplasms ,Lapatinib ,Neoplasms, Second Primary ,Trastuzumab ,Ado-Trastuzumab Emtansine ,Oncology ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Female ,Maytansine ,Biosimilar Pharmaceuticals - Abstract
Human epidermal growth factor receptor 2 (HER2)-targeted therapies improve survival for patients with HER2-positive breast cancer but carry risks of hematologic, cardiopulmonary, gastro-hepatobiliary, and other adverse events (AEs). In this review, we describe published AE incidences for HER2-targeted therapies for metastatic breast cancer (mBC).We searched PubMed and Embase to identify studies on HER2-targeted therapies in HER2-positive mBC, reporting on AEs of special interest, and published between January 1, 2009, and February 6, 2020. Treatment regimens were categorized into mutually exclusive therapy-based categories, with primary therapy determined by worldwide approval date.One hundred and fifty-three included articles assessed a combined 29,238 patients treated with the following therapy-based regimens: trastuzumab or biosimilars (78 studies), lapatinib (40), T-DM1 (ado-trastuzumab emtansine) (20), pertuzumab (14), neratinib (8), MM-302 (1), T-DXd (2), tucatinib (3), and pyrotinib (3). While direct comparisons of AE incidence are not warranted owing to study heterogeneity, proportions of patients experiencing any Grade 3 + AE ranged across therapy-based regimens from 39.4% (lapatinib) to 66.3% (neratinib). The most common hematologic AE of special interest, of any grade and regardless of causality, was leukopenia/white blood cells decreased [21.4% (T-DXd)-46.2% (pyrotinib)]. Cardiopulmonary AEs of special interest included interstitial lung disease [2.7% (trastuzumab)-5.2% (T-DXd)], pneumonitis [0.2% (lapatinib)-7.4% (trastuzumab)], and decreased ejection fraction [1% (T-DXd)-13.6% (trastuzumab)]. Gastro-hepatobiliary AEs of special interest included nausea [33.9% (trastuzumab)-78.3% (T-DXd)], vomiting [19.2% (T-DM1)-48.2% (T-DXd)], diarrhea [19.6% (T-DM1)-96.9% (pyrotinib)], and hepatotoxicity [5.9% (lapatinib)-53.6% (T-DM1)].Differing AE profiles for anti-HER2 therapies should be considered when assessing benefit-risk profile for treatment options.
- Published
- 2022
- Full Text
- View/download PDF
167. Praluzatamab Ravtansine, a CD166-Targeting Antibody- Drug Conjugate, in Patients with Advanced Solid Tumors: An Open-Label Phase I/II Trial
- Author
-
Valentina Boni, Mary J. Fidler, Hendrik-Tobias Arkenau, Alexander Spira, Funda Meric-Bernstam, Nataliya Uboha, Rachel E. Sanborn, Randy F. Sweis, Patricia LoRusso, Misako Nagasaka, Javier Garcia-Corbacho, Shadia Jalal, James J. Harding, Stella K. Kim, Iris H.C. Miedema, Danielle J. Vugts, Marc C. Huisman, Gerben J.C. Zwezerijnen, Guus A.M.S. van Dongen, C. Willemien Menke van der Houven van Oordt, Song Wang, Tam Dang, Ivan A. Zein, Olga Vasiljeva, Susan K. Lyman, Virginia Paton, Alison Hannah, Joyce F. Liu, Internal medicine, CCA - Imaging and biomarkers, Radiology and nuclear medicine, CCA - Cancer biology and immunology, AII - Inflammatory diseases, Amsterdam Neuroscience - Brain Imaging, and CCA - Cancer Treatment and quality of life
- Subjects
Cancer Research ,Immunoconjugates ,Oncology ,Neoplasms ,Tumor Microenvironment ,Humans ,Antineoplastic Agents ,Breast Neoplasms ,Female ,Maytansine - Abstract
Purpose: Praluzatamab ravtansine (CX-2009) is a conditionally activated Probody drug conjugate (PDC) comprising an anti-CD166 mAb conjugated to DM4, with a protease-cleavable linker and a peptide mask that limits target engagement in normal tissue and circulation. The tumor microenvironment is enriched for proteases capable of cleaving the linker, thereby releasing the mask, allowing for localized binding of CX-2009 to CD166. CX-2009 was evaluated in a phase I/II clinical trial for patients with advanced solid tumors. Patients and Methods: Eligible patients had metastatic cancer receiving ≥2 prior treatments. CX-2009 was administered at escalating doses every 3 weeks (0.25–10 mg/kg) or every 2 weeks (4–6 mg/kg). Primary objective was to determine the safety profile and recommended phase II dose (RP2D). Results: Of 99 patients enrolled, the most prevalent subtype was breast cancer (n = 45). Median number of prior therapies was 5 (range, 1–19). Dose-limiting toxicities were observed at 8 mg/kg every 3 weeks and 6 mg/kg every 2 weeks. On the basis of tolerability, the RP2D was 7 mg/kg every 3 weeks. Tumor regressions were observed at doses ≥4 mg/kg. In the hormone receptor–positive/HER2-nonamplified breast cancer subset (n = 22), 2 patients (9%) had confirmed partial responses, and 10 patients (45%) had stable disease. Imaging with zirconium-labeled CX-2009 confirmed uptake in tumor lesions and shielding of major organs. Activated, unmasked CX-2009 was measurable in 18 of 22 posttreatment biopsies. Conclusions: CD166 is a novel, ubiquitously expressed target. CX-2009 is the first conditionally activated antibody–drug conjugate to CD166 to demonstrate both translational and clinical activity in a variety of tumor types.
- Published
- 2022
- Full Text
- View/download PDF
168. Impacts of clinicopathological factors on efficacy of trastuzumab deruxtecan in patients with HER2-positive metastatic breast cancer
- Author
-
Hiromichi Nakajima, Kenichi Harano, Tokiko Nakai, Shota Kusuhara, Takehiro Nakao, Chikako Funasaka, Chihiro Kondoh, Nobuaki Matsubara, Yoichi Naito, Ako Hosono, Shuichi Mitsunaga, Genichiro Ishii, and Toru Mukohara
- Subjects
NA, not available ,Immunoconjugates ,PD, progressive disease ,IHC, immunohistochemical score ,Receptor, ErbB-2 ,hetero, heterogenous ,NLR, neutrophil-to-lymphocyte ratio ,Breast Neoplasms ,MT, mutant-type ,PR, partial response ,PLR, platelet-to-lymphocyte ratio ,Humans ,homo, homogenous ,Maytansine ,Trastuzumab deruxtecan ,amp, amplification ,HER2-Positive metastatic breast cancer ,skin and connective tissue diseases ,RC254-282 ,Retrospective Studies ,Ado-trastuzumab emtansine ,SD, stable disease ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Biomarker ,General Medicine ,Trastuzumab ,n, number ,WT, wild-type ,PFS, progression-free survival ,Camptothecin ,Female ,Original Article ,IDC, invasive ductal carcinoma ,Surgery - Abstract
Background The previous second-line treatment for HER2-positive metastatic breast cancer were ado-trastuzumab emtansine (T-DM1); however, its activity is decreased in tumors with heterogenous, reduced, or loss of HER2 expression. Trastuzumab deruxtecan (T-DXd) has recently been developed as a novel antibody-drug conjugate to overcome resistance to T-DM1. However, clinical evidence on its ability to overcome this resistance is limited. Materials and methods We retrospectively analyzed data for patients with HER2-positive metastatic breast cancer who received T-DXd at our institution from April 2020 to March 2021. We evaluated the associations between clinicopathological and molecular biomarkers and the efficacy of T-DXd. Results Twenty-two patients were enrolled in this study. The median progression-free survival (PFS) was 9.7 months (95% confidence interval [CI], 7.0–not reached [NR]), and the objective response rate (ORR) was 61.9%. The ORR and PFS were comparable between patients with HER2 immunohistochemistry scores of 3+ and 2+/1+ at initial diagnosis (ORR: 50.0% vs. 72.7%, p = 0.39; median PFS, 9.7 months [95%CI, 2.6–NR] vs. 8.3 months [95%CI, 7.1–NR]; hazard ratio, 1.86 [95%CI, 0.53–6.57], p = 0.34). Two patients with heterogenous HER2 expression had a partial response or long stable disease (≥6 months). Three of four patients with re-biopsy samples after anti-HER2 targeted therapy and with latest HER2 immunohistochemistry scores of 1+ experienced partial responses (75.0%) to T-DXd, but none had responded to prior T-DM1. Conclusions T-DXd demonstrated favorable activity in clinical practice. Moreover, T-DXd showed meaningful benefit in patients with heterogeneity, reduction, or loss of HER2 expression., Highlights • T-DXd showed favorable clinical activity against HER2-positive metastatic breast cancer. • T-DXd showed benefit in patients with heterogeneity, reduction, or loss of HER2 expression. • Patients with liver metastasis tended to have worse outcomes.
- Published
- 2022
- Full Text
- View/download PDF
169. Exploiting mesothelin in thymic carcinoma as a drug delivery target for anetumab ravtansine
- Author
-
Vincent Chen, Shigeki Umemura, Yumin Han, Renuka Raman, Robin Tucker, Joeffrey Chahine, In-Kyu Kim, Christoph Schatz, Sabine Zitzmann-Kolbe, Anette Sommer, Masanori Onda, Trevor Lee, Yongfeng He, and Giuseppe Giaccone
- Subjects
Cancer Research ,Immunoconjugates ,Thymoma ,Cell Survival ,Drug Synergism ,Thymus Neoplasms ,Xenograft Model Antitumor Assays ,Article ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Mice ,Oncology ,Cell Line, Tumor ,Mesothelin ,Animals ,Humans ,Female ,Maytansine ,Neoplasms, Glandular and Epithelial ,Cisplatin ,HT29 Cells ,Cell Proliferation - Abstract
BACKGROUND: Thymic epithelial tumours (TETs) are rare tumours comprised of thymomas and thymic carcinoma. Novel therapies are needed, especially in thymic carcinoma where the 5-year survival rate hovers at 30%. Mesothelin (MSLN), a surface glycoprotein that is cleaved to produce mature MSLN (mMSLN) and megakaryocyte potentiating factor (MPF), is expressed in limited tissues. However, its expression is present in various cancers, including thymic carcinoma, where it is expressed in 79% of cases. METHODS: We utilised flow cytometry, in vitro cytotoxicity assays, and an in vivo xenograft model in order to demonstrate the ability of the MSLN targeting antibody–drug conjugate (ADC) anetumab ravtansine (ARav) in inhibiting the growth of thymic carcinoma. RESULTS: Thymoma and thymic carcinoma cell lines express MSLN, and anetumab, the antibody moiety of ARav, was capable of binding MSLN expressing thymic carcinoma cells and internalising. ARav was effective at inhibiting the growth of thymic carcinoma cells stably transfected with mMSLN in vitro. In vivo, 15 mg/kg ARav inhibited T1889 xenograft tumour growth, while combining 7.5 mg/kg ARav with 4 mg/kg cisplatin yielded an additive effect on inhibiting tumour growth. CONCLUSIONS: These data demonstrate that anetumab ravtansine inhibits the growth of MSLN positive thymic carcinoma cells in vitro and in vivo.
- Published
- 2021
- Full Text
- View/download PDF
170. An intelligent cell-selective polymersome-DM1 nanotoxin toward triple negative breast cancer
- Author
-
Rainer Haag, Huanli Sun, Shujing Yue, Zhiyuan Zhong, and Yifan Zhang
- Subjects
Immunoconjugates ,Lung Neoplasms ,Mice, Nude ,Pharmaceutical Science ,Breast Neoplasms ,Triple Negative Breast Neoplasms ,Mice ,Breast cancer ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Maytansine ,Cytotoxicity ,Triple-negative breast cancer ,biology ,Chemistry ,Cancer ,Trastuzumab ,medicine.disease ,Hepatocellular carcinoma ,Polymersome ,Cancer research ,biology.protein ,Systemic administration ,Female ,Antibody - Abstract
Antibody-drug conjugates (ADCs) are among the most significant advances in clinical cancer treatments, however, they are haunted with fundamental issues like low drug/antibody ratio (DAR), need of large amount of antibody, and complex chemistry. Targeted nanomedicines while offering a promising alternative to ADCs are afflicted with drug leakage and inferior cancer-specificity. Herein, we developed an intelligent cell-selective nanotoxin based on anti-CD44 antibody-polymersome-DM1 conjugates (aCD44-AP-DM1) for potent treatment of solid tumors. DM1 was simultaneously coupled to vesicular membrane via disulfide bonds during self-assembly and anti-CD44 antibody was facilely clicked onto polymersome surface, tailor-making an optimal aCD44-AP-DM1 with a controlled antibody density of 5.0, extraordinary DAR of 275, zero drug leakage and rapid reduction-responsive DM1 release. aCD44-AP-DM1 displayed a high specificity and exceptional cytotoxicity toward MDA-MB-231 triple negative breast cancer, SMMC-7721 hepatocellular carcinoma and A549 non-small cell lung cancer cells with half-maximal inhibitory concentrations (IC50) of 21.4, 3.7 and 64.6 ng/mL, respectively, 3.6–47.2-fold exceeding non-targeted P-DM1. Intriguingly, the systemic administration of aCD44-AP-DM1 significantly suppressed subcutaneous MDA-MB-231 tumor xenografts in nude mice while intratumoral injection achieved complete tumor eradication in four out of five mice, without causing toxicity. This intelligent cell-selective nanotoxin has emerged as a better platform over ADCs for targeted cancer therapy.
- Published
- 2021
- Full Text
- View/download PDF
171. Indatuximab ravtansine plus dexamethasone with lenalidomide or pomalidomide in relapsed or refractory multiple myeloma: a multicentre, phase 1/2a study
- Author
-
Sagar Lonial, Afsaneh Abdolzade-Bavil, Asher Chanan-Khan, Markus Ruehle, Farima Barmaki-Rad, Nikhil C. Munshi, Suraj Chandwani, George Somlo, Kevin R. Kelly, Sundar Jagannath, Ashot Minasyan, Sumit Madan, Andrea Wartenberg-Demand, Eva Herrmann-Keiner, Todd M. Zimmerman, Leonard T. Heffner, Sikander Ailawadhi, Faiza Rharbaoui, Thomas Haeder, David S. Siegel, and Kenneth C. Anderson
- Subjects
Adult ,Male ,medicine.medical_specialty ,Immunoconjugates ,Maximum Tolerated Dose ,Angiogenesis Inhibitors ,Neutropenia ,Gastroenterology ,Dexamethasone ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,Maytansine ,Lenalidomide ,Multiple myeloma ,Aged ,Aged, 80 and over ,Bortezomib ,business.industry ,Hematology ,Middle Aged ,Pomalidomide ,medicine.disease ,Thalidomide ,Female ,Neoplasm Recurrence, Local ,Multiple Myeloma ,business ,Progressive disease ,medicine.drug - Abstract
Summary Background Indatuximab ravtansine (BT062) is an antibody-drug conjugate that binds to CD138 and synergistically enhances the antitumor activity of lenalidomide in preclinical models of multiple myeloma. This phase 1/2a study was done to determine the safety, activity, and pharmacokinetics of indatuximab ravtansine in combination with immunomodulatory drugs in patients with relapsed or refractory multiple myeloma. Methods This open-label, phase 1/2a study took place at nine hospital sites in the USA. Eligible patients were aged 18 years or older, had relapsed or refractory multiple myeloma, and ECOG performance status or Zubrod score of 2 or below. Patients who received indatuximab ravtansine with lenalidomide and dexamethasone (indatuximab ravtansine plus lenalidomide) had failure of at least one previous therapy. Patients treated with indatuximab ravtansine with pomalidomide and dexamethasone (indatuximab ravtansine plus pomalidomide) had failure of at least two previous therapies (including lenalidomide and bortezomib) and had progressive disease on or within 60 days of completion of their last treatment. In phase 1, patients received indatuximab ravtansine intravenously on days 1, 8, and 15 of each 28-day cycle in escalating dose levels of 80 mg/m2, 100 mg/m2, and 120 mg/m2, with lenalidomide (25 mg; days 1 to 21 every 28 days orally) and dexamethasone (20–40 mg; days 1, 8, 15, and 22 every 28 days). In phase 2, the recommended phase 2 dose of indatuximab ravtansine was given to an expanded cohort of patients in combination with lenalidomide and dexamethasone. The protocol was amended to allow additional patients to be treated with indatuximab ravtansine plus pomalidomide (4 mg; days 1 to 21 every 28 days orally) and dexamethasone, in a more heavily pretreated patient population than in the indatuximab ravtansine plus lenalidomide group. The phase 1 primary endpoint was to determine the dose-limiting toxicities and the maximum tolerated dose (recommended phase 2 dose) of indatuximab ravtansine, and the phase 2 primary endpoint was to describe the objective response rate (ORR; partial response or better) and clinical benefit response (ORR plus minor response). All patients were analysed for safety and all patients with post-treatment response assessments were analysed for activity. This study is registered with ClinicalTrials.gov , number NCT01638936 , and is complete. Findings 64 (86%) of 74 screened patients were enrolled between July 3, 2012, and June 30, 2015. 47 (73%) patients received indatuximab ravtansine plus lenalidomide (median follow-up 24·2 months [IQR 19·9–45·4]) and 17 (27%) received indatuximab ravtansine plus pomalidomide (24·1 months [17·7–36·7]). The maximum tolerated dose of indatuximab ravtansine plus lenalidomide was 100 mg/m2, and defined as the recommended phase 2 dose for indatuximab ravtansine plus pomalidomide. An objective response for indatuximab ravtansine plus lenalidomide was observed in 33 (71·7%) of 46 patients and in 12 (70·6%) of 17 patients in the indatuximab ravtansine plus pomalidomide group. The clinical benefit response for indatuximab ravtansine plus lenalidomide was 85% (39 of 46 patients) and for indatuximab ravtansine plus pomalidomide it was 88% (15 of 17 patients). The most common grade 3–4 adverse events in both groups were neutropenia (14 [22%] of 64 patients), anaemia (10 [16%]), and thrombocytopenia (seven [11%]). Treatment-emergent adverse events (TEAEs) that led to discontinuation occurred in 35 (55%) of the 64 patients. Five (8%) patients with a TEAE had a fatal outcome; none was reported as related to indatuximab ravtansine. Interpretation Indatuximab ravtansine in combination with immunomodulatory drugs shows preliminary antitumor activity, is tolerated, and could be further evaluated in patients with relapsed or refractory multiple myeloma. Funding Biotest AG.
- Published
- 2021
- Full Text
- View/download PDF
172. Neoadjuvant T-DM1/pertuzumab and paclitaxel/trastuzumab/pertuzumab for HER2+ breast cancer in the adaptively randomized I-SPY2 trial
- Author
-
Denise M. Wolf, Jane Perlmutter, Judy C. Boughey, A. Jo Chien, Meredith Buxton, Gillian L. Hirst, Douglas Yee, Angela DeMichele, Andres Forero-Torres, Scott M. Berry, Erin D. Ellis, Anthony D. Elias, Julia Wulfkuhle, Michael Alvarado, Christina Yau, Stacy L. Moulder, Nola M. Hylton, Rita Nanda, Amy Wilson, Adam Asare, Debu Tripathy, Claudine Isaacs, Melissa Paoloni, Rosa I. Gallagher, Laura J. Esserman, Richard Schwab, W. Fraser Symmans, Cheryl Ewing, Laura J. van't Veer, Jeffrey B. Matthews, Teresa Helsten, Julia L. Clennell, Barbara Haley, Emanuel F. Petricoin, Katherine Steeg, Smita Asare, Ashish Sanil, Rachel L. Yung, Erin P. Crane, Erin Roesch, Hyo S. Han, Ruby Singhrao, Michelle E. Melisko, Hope S. Rugo, Kathy S. Albain, Donald A. Berry, Anne M. Wallace, Julie E. Lang, Amy S. Clark, Kathleen Kemmer, and Lamorna Brown-Swigart
- Subjects
Oncology ,Receptor, ErbB-2 ,General Physics and Astronomy ,Ado-Trastuzumab Emtansine ,chemistry.chemical_compound ,Breast cancer ,ErbB-2 ,Trastuzumab ,Monoclonal ,skin and connective tissue diseases ,Humanized ,Cancer ,Multidisciplinary ,Tumor ,medicine.diagnostic_test ,Middle Aged ,Neoadjuvant Therapy ,Paclitaxel ,6.1 Pharmaceuticals ,Pertuzumab ,medicine.drug ,Receptor ,Adult ,medicine.medical_specialty ,Cyclophosphamide ,Science ,Clinical Trials and Supportive Activities ,Context (language use) ,Breast Neoplasms ,Antibodies, Monoclonal, Humanized ,Article ,General Biochemistry, Genetics and Molecular Biology ,Antibodies ,Clinical Research ,Internal medicine ,Biopsy ,Breast Cancer ,Biomarkers, Tumor ,medicine ,Humans ,Doxorubicin ,Maytansine ,neoplasms ,Aged ,business.industry ,Evaluation of treatments and therapeutic interventions ,General Chemistry ,Translational research ,medicine.disease ,chemistry ,business ,Biomarkers - Abstract
HER2-targeted therapy dramatically improves outcomes in early breast cancer. Here we report the results of two HER2-targeted combinations in the neoadjuvant I-SPY2 phase 2 adaptive platform trial for early breast cancer at high risk of recurrence: ado-trastuzumab emtansine plus pertuzumab (T-DM1/P) and paclitaxel, trastuzumab and pertuzumab (THP). Eligible women have >2.5 cm clinical stage II/III HER2+ breast cancer, adaptively randomized to T-DM1/P, THP, or a common control arm of paclitaxel/trastuzumab (TH), followed by doxorubicin/cyclophosphamide, then surgery. Both T-DM1/P and THP arms ‘graduate’ in all subtypes: predicted pCR rates are 63%, 72% and 33% for T-DM1/P (n = 52), THP (n = 45) and TH (n = 31) respectively. Toxicity burden is similar between arms. Degree of HER2 pathway signaling and phosphorylation in pretreatment biopsy specimens are associated with response to both T-DM1/P and THP and can further identify highly responsive HER2+ tumors to HER2-directed therapy. This may help identify patients who can safely de-escalate cytotoxic chemotherapy without compromising excellent outcome., HER2-targeted therapy improves patient’s outcome in early breast cancer. Here, the authors present the efficacy and biomarker analysis of two HER2-targeted combinations (ado-trastuzumab emtansine plus pertuzumab and paclitaxel, trastuzumab and pertuzumab) in the context of the neoadjuvant I-SPY2 phase 2 adaptive platform trial for early breast cancer at high risk of recurrence.
- Published
- 2021
173. Trastuzumab does not bind rat or mouse ErbB2/neu: implications for selection of non-clinical safety models for trastuzumab-based therapeutics
- Author
-
Jun Guo, Noel Dybdal, Ben-Quan Shen, Daniela Bumbaca Yadav, Gail Lewis Phillips, William R Proctor, and James R. Kiefer
- Subjects
Cancer Research ,Receptor, ErbB-2 ,T-DM1 ,Breast Neoplasms ,Antibodies, Monoclonal, Humanized ,chemistry.chemical_compound ,Mice ,Preclinical Study ,Trastuzumab ,Tandem Mass Spectrometry ,medicine ,Animals ,Humans ,Maytansine ,Viability assay ,skin and connective tissue diseases ,neoplasms ,Trastuzumab emtansine ,chemistry.chemical_classification ,Rodent ,biology ,Catabolism ,Chemistry ,Binding ,Amino acid ,Rats ,Oncology ,Cell culture ,biology.protein ,Cancer research ,Immunohistochemistry ,Female ,Antibody ,medicine.drug ,Human ,Chromatography, Liquid - Abstract
Purpose Assessment of non-clinical safety signals relies on understanding species selectivity of antibodies. This is particularly important with antibody–drug conjugates, where it is key to determine target-dependent versus target-independent toxicity. Although it appears to be widely accepted that trastuzumab does not bind mouse or rat HER2/ErbB2/neu, numerous investigators continue to use mouse models to investigate safety signals of trastuzumab and trastuzumab emtansine (T-DM1). We, therefore, conducted a broad array of both binding and biologic studies to demonstrate selectivity of trastuzumab for human HER2 versus mouse/rat neu. Methods Binding of anti-neu and anti-HER2 antibodies was assessed by ELISA, FACS, IHC, Scatchard, and immunoblot methods in human, rat, and mouse cell lines. In human hepatocytes, T-DM1 uptake and catabolism were measured by LC-MS/MS; cell viability changes were determined using CellTiter-Glo. Results Our data demonstrate, using different binding methods, lack of trastuzumab binding to rat or mouse neu. Structural studies show important amino acid differences in the trastuzumab-HER2 binding interface between mouse/rat and human HER2 ECD. Substitution of these rodent amino acid residues into human HER2 abolish binding of trastuzumab. Cell viability changes, uptake, and catabolism of T-DM1 versus a DM1 non-targeted control ADC were comparable, indicating target-independent effects of the DM1-containing ADCs. Moreover, trastuzumab binding to human or mouse hepatocytes was not detected. Conclusions These data, in total, demonstrate that trastuzumab, and by extension T-DM1, do not bind rat or mouse neu, underscoring the importance of species selection for safety studies investigating trastuzumab or trastuzumab-based therapeutics.
- Published
- 2021
174. HPLC-DAD validated method for DM4 and its metabolite S-Me-DM4 quantification in biological matrix for clinical and pharmaceutical applications.
- Author
-
Lovato G, Ciriolo L, Perrucci M, Federici L, Ippoliti R, Iacobelli S, Capone E, Locatelli M, and Sala G
- Subjects
- Humans, Reproducibility of Results, Chromatography, High Pressure Liquid methods, Pharmaceutical Preparations, Maytansine
- Abstract
The present study focuses on the development and validation of an HPLC-DAD methodology for the detection of a potent chemotherapeutic agent, Maytansinoid Ravtansine (DM4), and its metabolite, S-methyl-DM4 (S-Me-DM4), in plasma samples. Methodologically, after a simple protein precipitation with acetonitrile and after drying 1 mL of supernatant, the sample (suspended with N,N-Dimethylacetamide, DMA) was directly analyzed by HPLC under isocratic elution using a mobile phase comprising milliQ water and methanol (25:75, v:v), both acidified with 0.1 % v:v formic acid. Employing a flow rate of 1.0 mL/min and a reversed-phase GraceSmart RP18 column thermostated at 40 °C, we achieved complete resolution and separation of DM4 and S-Me-DM4 within 13 min. The optimized injection volume of 20 μL and the wavelength set at 254 nm were utilized for quantitative analyses. Rigorous validation has not only ensured its reliability and reproducibility but has also addressed potential limitations associated with methodological inconsistency. The limit of detection and quantification of the method were 0.025 and 0.06 μg/mL for both the analytes, respectively. The calibration curve showed a good linearity in the range 0.06-20 μg/mL. For both analytes, the intraday precision and trueness were 2.3-8.2 % and -1.1 to 3.1 %, respectively, while the interday values were 0.7-10.1 % and -10.4 to 7.5 %, respectively. The developed methodology enables the concurrent determination and quantification of free DM4 and its metabolite, free S-Me-DM4, making it a valuable tool for assessing the pharmacokinetics and pharmacodynamics of DM4-based therapies. In addition, the procedure was successfully applied to analyse the presence of free DM4 or its metabolite, free S-Me-DM4, in human plasma samples spiked with the 1959-sss/DM4 antibody-drug conjugate (ADC). The utilization of the herein validated methodology allowed to confirm the presence of these analytes, thereby providing insights into their potential release from the ADC structure., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
175. Affibody-derived drug conjugates: Potent cytotoxic molecules for treatment of HER2 over-expressing tumors.
- Author
-
Altai, Mohamed, Liu, Hao, Ding, Haozhong, Mitran, Bogdan, Edqvist, Per-Henrik, Tolmachev, Vladimir, Orlova, Anna, and Gräslund, Torbjörn
- Subjects
- *
ANTIBODY-drug conjugates , *TUMOR treatment , *ANTINEOPLASTIC agents , *HER2 protein , *MONOMERS - Abstract
Abstract Patients with HER2-positive tumors often suffer resistance to therapy, warranting development of novel treatment modalities. Affibody molecules are small affinity proteins which can be engineered to bind to desired targets. They have in recent years been found to allow precise targeting of cancer specific molecular signatures such as the HER2 receptor. In this study, we have investigated the potential of an affibody molecule targeting HER2, Z HER2:2891 , conjugated with the cytotoxic maytansine derivate MC-DM1, for targeted cancer therapy. Z HER2:2891 was expressed as a monomer (Z HER2:2891), dimer ((Z HER2:2891) 2) and dimer with an albumin binding domain (ABD) for half-life extension ((Z HER2:2891) 2 -ABD). All proteins had a unique C-terminal cysteine that could be used for efficient and site-specific conjugation with MC-DM1. The resulting affibody drug conjugates were potent cytotoxic molecules for human cells over-expressing HER2, with sub-nanomolar IC 50 -values similar to trastuzumab emtansine, and did not affect cells with low HER2 expression. A biodistribution study of a radiolabeled version of (Z HER2:2891) 2 -ABD-MC-DM1, showed that it was taken up by the tumor. The major site of off-target uptake was the kidneys and to some extent the liver. (Z HER2:2891) 2 -ABD-MC-DM1 was found to have a half-life in circulation of 14 h. The compound was tolerated well by mice at 8.5 mg/kg and was shown to extend survival of mice bearing HER2 over-expressing tumors. The findings in this study show that affibody molecules are a promising class of engineered affinity proteins to specifically deliver small molecular drugs to cancer cells and that such conjugates are potential candidates for clinical evaluation on HER2-overexpressing cancers. Graphical abstract Unlabelled Image [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
176. Antibody drug conjugates of cleavable amino-alkyl and aryl maytansinoids.
- Author
-
Nittoli, Thomas, Kelly, Marcus P., Delfino, Frank, Rudge, John, Kunz, Arthur, Markotan, Thomas, Spink, Jan, Chen, Zhaoyuan, Shan, Jing, Navarro, Elizabeth, Tait, Michele, Provoncha, Kathleen, Giurleo, Jason, Zhao, Feng, Jiang, Xiaobo, Hylton, Donna, Makonnen, Sosina, Hickey, Carlos, Kirshner, Jessica R., and Thurston, Gavin
- Subjects
- *
THERAPEUTIC use of immunoglobulins , *NATURAL products , *ANTINEOPLASTIC agents , *MAYTANSINE , *ENDOTHELIAL growth factors - Abstract
Natural products have been used for many medicinal purposes for centuries. Antibody drug conjugates (ADCs) have utilized this rich source of small molecule therapeutics to produce several clinically useful treatments. ADCs based on the natural product maytansine have been successful clinically. The authors further the utility of the anti-cancer natural product maytansine by developing efficacious payloads and linker-payloads for conjugating to antibodies. The success of our approach was realized in the EGFRvIII targeting ADC EGFRvIII-16. The ADC was able to regress tumors in 2 tumor models (U251/EGFRvIII and MMT/EGFRvIII). When compared to a positive control ADC, the efficacy observed was similar or improved while the isotype control ADCs had no effect. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
177. C3 ester side chain plays a pivotal role in the antitumor activity of Maytansinoids
- Author
-
Anjie Xia, Jinliang Yang, Yuxi Wang, Minhao Huang, Lun Tan, Wenting Li, Yuyan Li, and Zhixiong Zhang
- Subjects
Models, Molecular ,0301 basic medicine ,Immunoconjugates ,Swine ,Stereochemistry ,Biophysics ,Antineoplastic Agents ,Maytansinoid ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Tubulin ,Drug Discovery ,Side chain ,Animals ,Humans ,Moiety ,Maytansine ,Molecular Biology ,biology ,Esters ,Cell Biology ,Ligand (biochemistry) ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Chirality (chemistry) ,Methyl group ,Conjugate - Abstract
Maytansinoids, the chemical derivatives of Maytansine, are commonly used as potent cytotoxic payloads in antibody-drug conjugates (ADC). Structure-activity-relationship studies had identified the C3 ester side chain as a critical element for antitumor activity of maytansinoids. The maytansinoids bearing the methyl group at C3 position with D configuration were about 100 to 400-fold less cytotoxic than their corresponding L-epimers toward various cell lines. The detailed mechanism of how chirality affects the anticancer activity remains elusive. In this study, we determined the high-resolution crystal structure of tubulin in complex with maytansinol, L-DM1-SMe and D-DM1-SMe. And we found the carbonyl oxygen atom of the ester moiety and the tail thiomethyl group at C3 side chain of L-DM1-SMe form strong intramolecular interaction with the hydroxyl at position 9 and the benzene ring, respectively, fixing the bioactive conformation and enhancing the binding affinity. Additionally, ligand-based and structure-based virtually screening methods were used to screen the commercially macrocyclic compounds library, and 15 macrocyclic structures were picketed out as putatively new maytansine-site inhibitors. Our study provides a possible strategy for the rational discovery of next-generation maytansine site inhibitors.
- Published
- 2021
- Full Text
- View/download PDF
178. New insights into the anticancer therapeutic potential of maytansine and its derivatives.
- Author
-
Zafar, Sameen, Armaghan, Muhammad, Khan, Khushbukhat, Hassan, Nazia, Sharifi-Rad, Javad, Habtemariam, Solomon, Kieliszek, Marek, Butnariu, Monica, Bagiu, Iulia-Cristina, Bagiu, Radu Vasile, and Cho, William C.
- Subjects
- *
TUBULINS , *CELL cycle , *MONOCLONAL antibodies , *MICROTUBULES , *CLINICAL medicine , *ANTINEOPLASTIC agents - Abstract
Maytansine is a pharmacologically active 19-membered ansamacrolide derived from various medicinal plants and microorganisms. Among the most studied pharmacological activities of maytansine over the past few decades are anticancer and anti-bacterial effects. The anticancer mechanism of action is primarily mediated through interaction with the tubulin thereby inhibiting the assembly of microtubules. This ultimately leads to decreased stability of microtubule dynamics and cause cell cycle arrest, resulting in apoptosis. Despite its potent pharmacological effects, the therapeutic applications of maytansine in clinical medicine are quite limited due to its non-selective cytotoxicity. To overcome these limitations, several derivatives have been designed and developed mostly by modifying the parent structural skeleton of maytansine. These structural derivatives exhibit improved pharmacological activities as compared to maytansine. The present review provides a valuable insight into maytansine and its synthetic derivatives as anticancer agents. [Display omitted] • Maytansine is a naturally occurring microtubule destabilizing agent. • High systemic toxicity of maytansine limited its therapeutic window. • Development of synthetic derivatives has enhanced the therapeutic potential of the parent compound. • Conjugation of monoclonal antibodies with maytansine derivatives have increased their efficiency against various types of cancer. • T-DM1, is an FDA approved ADC targeting the Her2 + breast cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
179. New Cancer Gene Therapy Data Have Been Reported by Researchers at Sichuan University (Biomimetic Nanoerythrosome-coated Aptamer-dna Tetrahedron/maytansine Conjugates: Ph-responsive and Targeted Cytotoxicity for Her2-positive Breast Cancer).
- Subjects
HER2 positive breast cancer ,CANCER genes ,GENE therapy ,CANCER treatment ,RESEARCH personnel - Abstract
Researchers at Sichuan University in Chengdu, China have reported new data on cancer gene therapy. The study focuses on the use of DNA materials as nanocarriers for targeted cancer therapy. The researchers developed a drug delivery system using an HER2-targeted DNA-aptamer-modified DNA tetrahedron combined with maytansine (DM1). They also applied a biomimetic camouflage to enhance drug delivery and tumor-stimulated drug release. The hybrid erythrosome-based nanoparticles showed promising results in inhibiting HER2-positive cancer. This research represents an important step in the development of DNA-based medicine and biomimetic cell membrane materials for cancer treatment. [Extracted from the article]
- Published
- 2023
180. Effectiveness and cost-effectiveness of trastuzumab emtansine in women with HER2-positive locally advanced or metastatic breast cancer: A systematic review and meta-analysis
- Author
-
Yu Ko, Yingchih Yeh, and Chiehfeng Chen
- Subjects
Immunoconjugates ,Cytotoxins ,Receptor, ErbB-2 ,Cost-Benefit Analysis ,Breast Neoplasms ,General Medicine ,Trastuzumab ,Ado-Trastuzumab Emtansine ,Antibodies, Monoclonal, Humanized ,Disease-Free Survival ,Oncology ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Radiology, Nuclear Medicine and imaging ,Female ,Maytansine - Abstract
Trastuzumab emtansine (T-DM1) is a human epidermal growth factor receptor-2 targeted antibody-drug conjugate that contains a monoclonal antibody, trastuzumab, covalently linked to DM1, a small molecule cytotoxin.We conducted a systematic review and meta-analysis of published trials to examine the efficacy and safety of T-DM1 for patients with HER2-positive metastatic breast cancer. In addition, we systematically reviewed existing economic evaluations of T-DM1. An electronic literature search of online databases (Medline, CENTRAL, and Embase) was performed. Randomized controlled trials that compared T-DM1 with other active treatment agents were eligible for inclusion. In addition, studies that involved T-DM1 as one of the treatment comparators in an economic evaluation were included. Four trials with a total of 2462 participants were included in this meta-analysis.Pooled results showed T-DM1 substantially improved overall survival (hazard ratio [HR], 0.75; 95% confidence interval [CI], 0.67-0.85; IT-DM1 is effective for the treatment of patients with HER2-positive metastatic breast cancer, and it demonstrates a tolerable safety profile compared with other active controls. Little evidence was available regarding the cost-effectiveness of T-DM1 so no conclusions can be drawn.
- Published
- 2022
181. Effect of Ado-Trastuzumab Emtansine on Autologous Platelet Kinetics and Function
- Author
-
Ali M. Ansary, Moritz Stolla, Jill Corson, Arianne Cundy, S. Lawrence Bailey, Esther Pellham, Morgan Bawcom-Randall, Sherrill J. Slichter, and Vijayakrishna K. Gadi
- Subjects
Blood Platelets ,Cancer Research ,Receptor, ErbB-2 ,Breast Neoplasms ,Trastuzumab ,Ado-Trastuzumab Emtansine ,Antibodies, Monoclonal, Humanized ,Indium ,Thrombocytopenia ,Kinetics ,Oncology ,Humans ,Female ,Maytansine - Abstract
PURPOSE Ado-trastuzumab emtansine (T-DM1) treatment results in grade 3-4 thrombocytopenia in 8%-13% of patients. Prior in vitro studies reported T-DM1 inhibition of megakaryocyte maturation as the cause of decreased platelet production. The current observational study was initiated to evaluate causes of thrombocytopenia in patients with metastatic breast cancer. MATERIALS AND METHODS Patients with human epidermal growth factor receptor 2–positive metastatic breast cancer (N = 11) were enrolled in this postmarket safety study. 111-Indium- radiolabeled autologous platelet recoveries and survivals as well as serial platelet counts, bleeding time assays, and platelet aggregation responses to a wide range of agonists were performed at baseline (BL) and during two consecutive cycles of the drug (3.6 mg/kg IV once every 3 weeks). RESULTS Platelet nadirs occurred earlier in cycle 2 than in cycle 1. Average nadir counts (% BL) in cycles 1 and 2 were 116,000/µL (53% ± 6%) and 115,000/µL (51% ± 9%), respectively, with return to BL by D15 in both cycles. BL platelet survival averaged 8.8 (± 0.3) days but progressively shortened to 5.5 (± 0.5) days during cycle 1 and to 4.6 (± 0.3) days during cycle 2 ( P < .001 compared with BL for both cycles). Aggregation responses to all agonists decreased during the study, both in cycle 1 and cycle 2. CONCLUSION Following T-DM1 administration, we observed statistically significant progressive decreases in platelet survivals and decreased platelet function from BL values. In distinction to published in vitro studies, these unexpected results indicate a direct toxic effect of T-DM1 on patients' autologous circulating platelets.
- Published
- 2022
- Full Text
- View/download PDF
182. A Novel Antibody-Drug Conjugate Targeting Nectin-2 Suppresses Ovarian Cancer Progression in Mouse Xenograft Models
- Author
-
Yun Hee Sim, Yun Jung Um, Jeong-Yang Park, Min-Duk Seo, and Sang Gyu Park
- Subjects
Ovarian Neoplasms ,Immunoconjugates ,Organic Chemistry ,nectin-2 ,ovarian cancer ,chimeric antibody ,antibody-drug conjugate ,General Medicine ,Poly(ADP-ribose) Polymerase Inhibitors ,Carcinoma, Ovarian Epithelial ,Xenograft Model Antitumor Assays ,Catalysis ,Computer Science Applications ,Inorganic Chemistry ,Mice ,Cell Line, Tumor ,Immunoglobulin G ,Humans ,Animals ,Heterografts ,Female ,Maytansine ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy ,Platinum ,Cell Proliferation - Abstract
Ovarian cancer is the fifth leading cause of cancer, followed by front line is mostly platinum agents and PARP inhibitors, and very limited option in later lines. Therefore, there is a need for alternative therapeutic options. Nectin-2, which is overexpressed in ovarian cancer, is a known immune checkpoint that deregulates immune cell function. In this study, we generated a novel anti-nectin-2 antibody (chimeric 12G1, c12G1), and further characterized it using epitope mapping, enzyme-linked immunosorbent assay, surface plasmon resonance, fluorescence-activated cell sorting, and internalization assays. The c12G1 antibody specifically bound to the C2 domain of human nectin-2 with high affinity (KD = 2.90 × 10−10 M), but not to mouse nectin-2. We then generated an antibody-drug conjugate comprising the c12G1 antibody conjugated to DM1 and investigated its cytotoxic effects against cancer cells in vitro and in vivo. c12G1-DM1 induced cell cycle arrest at the mitotic phase in nectin-2-positive ovarian cancer cells, but not in nectin-2-negative cancer cells. c12G1-DM1 induced ~100-fold cytotoxicity in ovarian cancer cells, with an IC50 in the range of 0.1 nM~7.4 nM, compared to normal IgG-DM1. In addition, c12G1-DM1 showed ~91% tumor growth inhibition in mouse xenograft models transplanted with OV-90 cells. These results suggest that c12G1-DM1 could be used as a potential therapeutic agent against nectin-2-positive ovarian cancers.
- Published
- 2022
183. Ado-trastuzumab emtansine in the treatment of lung adenocarcinoma with ERBB2 mutation: a case report and literature review
- Author
-
Hao Wang, Yang He, Weipeng Zhao, and Zhongsheng Tong
- Subjects
Pharmacology ,Male ,Cancer Research ,Lung Neoplasms ,Receptor, ErbB-2 ,Adenocarcinoma of Lung ,Breast Neoplasms ,Trastuzumab ,Ado-Trastuzumab Emtansine ,Oncology ,Carcinoma, Non-Small-Cell Lung ,Mutation ,Humans ,Pharmacology (medical) ,Maytansine ,Aged - Abstract
The erb-b2 receptor tyrosine kinase 2 (ERBB2), also known as HER2, has long been recognized as an oncogenic driver in some breast and gastroesophageal cancers, and ERBB2-targeted therapies are standard for ERBB2-positive breast and gastric cancer. However, there are currently no standard therapies targeting the ERBB2 pathway in non-small cell lung cancer. Recently, somatic mutations in ERBB2 have been reported in 2-3% of patients with advanced lung adenocarcinoma, these mutations are trans-forming in lung cancer models and result in kinase activation, conferring some in-vitro sensitivity to trastuzumab. The ado-trastuzumab emtansine (T-DM1) is an antibody-drug conjugate composed of trastuzumab joined via a stable linker to DM1. In this report, a 67-year-old male patient was diagnosed with advanced lung adenocarcinoma with multiple lymph node metastases, and multi-chemotherapy and immunotherapy were not effective. The results of genetic testing indicated a non-frameshift insertion mutation in exon 20 of the ERBB2 gene. The patients received T-DM1 at a dose of 3.6 mg/kg by intravenous infusion every 21 days until for 12 cycles. Partial response appeared in the tumor lesions after treatment for four cycles, and PET-computer tomography showed the tumor lesions were effectively controlled, and the efficacy evaluation was complete response after treatment for six cycles. Although the patient experienced second degree of thrombocytopenia during the treatment, the corresponding symptomatic treatment was taken, and the platelets could return to normal before the next cycle of T-DM1. Follow-up review showed the patient is in good health and the tumor has not recurred.
- Published
- 2022
184. Activity of trastuzumab emtansine (T-DM1) in 3D cell culture
- Author
-
Robert Kraft, Jean Zheng Boyer, Eslie Dennis, Gail Lewis Phillips, Penny Towne, Elizabeth Vela, Karl Garsha, Brittany Admire, and Hiro Nitta
- Subjects
0301 basic medicine ,Cancer Research ,Receptor, ErbB-2 ,media_common.quotation_subject ,T-DM1 ,Cell ,Cell Culture Techniques ,Breast Neoplasms ,Drug resistance ,Ado-Trastuzumab Emtansine ,03 medical and health sciences ,3D cell culture ,chemistry.chemical_compound ,0302 clinical medicine ,Preclinical Study ,In vivo ,Cell Line, Tumor ,medicine ,Humans ,Maytansine ,Viability assay ,Internalization ,Drug efficacy study ,media_common ,Chemistry ,Trastuzumab ,Breast cancer cell lines ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Cell culture ,Trastuzumab emtansine ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Heterogeneity - Abstract
Background Cell spheroids and aggregates generated from three-dimensional (3D) cell culture methods are similar to in vivo tumors in terms of tissue morphology, biology, and gene expression, unlike cells grown in 2D cell cultures. Breast cancer heterogeneity is one of the main drug resistant mechanisms and needs to be overcome in order to increase the efficacy of drug activity in its treatments. Methods We performed a unique 3D cell culture and drug efficacy study with trastuzumab emtansine (Kadcyla®, T-DM1) across five breast cancer cell lines (BT-474, SK-BR-3, MDA-MB-361, MDA-MB-175, and MCF-7) that were previously investigated in 2D cell culture. We performed HER2 IHC staining, cell viability experiments, Gene-protein-assay (GPA), and T-DM1 internalization studies. Results We obtained significantly different results including higher IC50 for some of the cell lines. Our GPA showed some significant heterogeneous HER2 gene and protein expression in 3D cultured spheroids or aggregates. The fluorescent images also showed that a longer incubation time is needed for T-DM1 to be internalized effectively into 3D cultured spheroids or aggregates. Conclusion Our study demonstrated that the difference of T-DM1 drug activity in 3D spheroids or aggregates might be due to tumor heterogeneity and less efficient internalization of T-DM1 that is not seen using 2D cell culture models. Drug studies using 3D cell culture are expected to provide biologically relevant models for determining drug activity in tumor tissue in future drug response and resistance research.
- Published
- 2021
185. Trastuzumab-emtansine induced pleural and pericardial effusions
- Author
-
Sandrine Combret, Aurélie Grandvuillemin, Charlène Boulay, Pauline Lory, Didier Mayeur, Antonin Schmitt, Nils Martin, and Jeffrey Lombardi
- Subjects
musculoskeletal diseases ,Receptor, ErbB-2 ,medicine.drug_class ,Breast Neoplasms ,Ado-Trastuzumab Emtansine ,Antibodies, Monoclonal, Humanized ,Monoclonal antibody ,Pericardial effusion ,Pericardial Effusion ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Trastuzumab ,medicine ,Humans ,Maytansine ,Pharmacology (medical) ,business.industry ,Human epidermal growth factor ,medicine.disease ,Oncology ,chemistry ,Trastuzumab emtansine ,030220 oncology & carcinogenesis ,Cancer research ,Female ,business ,030215 immunology ,medicine.drug ,Conjugate - Abstract
Introduction Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate which combine trastuzumab (T), a monoclonal antibody targeting the human epidermal growth factor receptor-2 (HER2), and a cytotoxic molecule derived from maytansine (DM1). Case report We report the first case of T-DM1-associated pleural and pericardial effusions three weeks after the second course of T-DM1 in a patient with breast cancer. Drug-induced pleural and pericardial effusions was implicated in the absence of other etiologies. The Naranjo Scale indicated a probable drug-induced adverse reaction. Management & outcome: The patient fully recovered after thoracentesis and discontinuation of T-DM1. The patient has reported no side effect after the sixth course of trastuzumab. Discussion To our knowledge, this is the first case in the literature of bilateral pleural and pericardial effusions in a patient treated with T-DM1. The successful initiation of treatment with trastuzumab following withdrawal of T-DM1 suggests that emtansine played a role in the development of bilateral pleural and pericardial effusions. We hypothesize that the patient’s condition was a result of a local inflammatory reaction to emtansine by direct toxicity.
- Published
- 2021
- Full Text
- View/download PDF
186. Trastuzumab emtansine (T-DM1) versus trastuzumab in Chinese patients with residual invasive disease after neoadjuvant chemotherapy and HER2-targeted therapy for HER2-positive breast cancer in the phase 3 KATHERINE study
- Author
-
Chiun-Sheng Huang, Guofang Sun, Ling-Ming Tseng, Yi Feng, Kunwei Shen, Youngsen Yang, Ava Kwong, Shusen Wang, Mei-Ching Liu, Ting-Ying Ng, Zhimin Shao, Shin-Cheh Chen, and Iris Renfei Yan
- Subjects
musculoskeletal diseases ,0301 basic medicine ,Oncology ,China ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,medicine.medical_specialty ,Receptor, ErbB-2 ,medicine.medical_treatment ,Breast Neoplasms ,Ado-Trastuzumab Emtansine ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,Trastuzumab ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Clinical endpoint ,Humans ,Medicine ,Maytansine ,skin and connective tissue diseases ,Adverse effect ,Chemotherapy ,Taxane ,business.industry ,medicine.disease ,Neoadjuvant Therapy ,Discontinuation ,030104 developmental biology ,chemistry ,Trastuzumab emtansine ,030220 oncology & carcinogenesis ,Female ,business ,medicine.drug - Abstract
In the KATHERINE study (NCT01772472), patients with HER2-positive early breast cancer (EBC) and residual invasive disease after neoadjuvant chemotherapy plus HER2-targeted therapy who were treated with adjuvant trastuzumab emtansine (T-DM1) had a 50% reduction in the risk of an invasive disease-free survival (IDFS) event compared to patients treated with adjuvant trastuzumab. In metastatic disease, T-DM1 has resulted in higher rates of thrombocytopenia in Asian versus non-Asian patients. Here, we report safety and efficacy in Chinese patients from KATHERINE. Patients with HER2-positive EBC and residual invasive disease after taxane- and trastuzumab-containing neoadjuvant chemotherapy followed by surgery were randomized 1:1 to 14 cycles of adjuvant T-DM1 or trastuzumab. The primary endpoint was time to an IDFS event. Among Chinese patients (T-DM1 n = 51, trastuzumab n = 50), T-DM1 treatment resulted in a 43% reduction in risk of an IDFS event compared to trastuzumab (HR = 0.57; 95% CI 0.25–1.31), with similar results for secondary endpoints. As in the global population, Chinese patients receiving T-DM1 versus trastuzumab had more grade ≥ 3 adverse events (AEs; 39.2% versus 4.1%) and AEs leading to treatment discontinuation (27.5% versus 0%). The most common grade ≥ 3 AE with T-DM1 was thrombocytopenia (21.6%), a frequency higher than the frequency in the global population (5.7%). Grade ≥ 3 hemorrhage was reported in 1 patient (T-DM1 arm). In the KATHERINE study, T-DM1 demonstrated increased efficacy compared to trastuzumab in Chinese patients. Consistent with previous data in Asian patients, T-DM1 was associated with more grade ≥ 3 AEs, and AEs leading to discontinuation, which was driven by an increase in thrombocytopenia.
- Published
- 2021
- Full Text
- View/download PDF
187. Improved AP-3 production through combined ARTP mutagenesis, fermentation optimization, and subsequent genome shuffling
- Author
-
Qiang Hua, Juan Li, Fengxian Hu, and Siyu Guo
- Subjects
0106 biological sciences ,0301 basic medicine ,Genome shuffling ,Mutant ,Mutagenesis (molecular biology technique) ,Bioengineering ,01 natural sciences ,Applied Microbiology and Biotechnology ,law.invention ,Plasma ,03 medical and health sciences ,Bacterial Proteins ,law ,010608 biotechnology ,Gene expression ,Maytansine ,Food science ,Strain (chemistry) ,Chemistry ,DNA Shuffling ,General Medicine ,Actinobacteria ,030104 developmental biology ,Metabolic Engineering ,Batch Cell Culture Techniques ,Mutagenesis ,Fermentation ,Recombinant DNA ,Actinosynnema pretiosum ,Biotechnology - Abstract
Ansamitocin (AP-3) is an ansamycins antibiotic isolated from Actinosynnema pretiosum and demonstrating high anti-tumor activity. To improve AP-3 production, the A. pretiosum ATCC 31565 strain was treated with atmospheric and room temperature plasma (ARTP). Four stable mutants were obtained by ARTP, of which the A. pretiosum L-40 mutant produced 242.9 mg/L AP-3, representing a 22.5% increase compared to the original wild type strain. With seed medium optimization, AP-3 production of mutant L-40 reached 307.8 mg/L; qRT-PCR analysis revealed that AP-3 biosynthesis-related gene expression was significantly up-regulated under optimized conditions. To further improve the AP-3 production, genome shuffling (GS) technology was used on the four A. pretiosum mutants by ARTP. After three rounds of GS combined with high-throughput screening, the genetically stable recombinant strain G3-96 was obtained. The production of AP-3 in the G3-96 strain was 410.1 mg/L in shake flask cultures, which was 44.5% higher than the L-40 production from the parental strain, and AP-3 was increased by 93.8% compared to the wild-type A. pretiosum. These results suggest that the combination of mutagenesis, seed medium optimization, and GS technology can effectively improve the AP-3 production capacity of A. pretiosum and provide an enabling methodology for AP-3 industrial production.
- Published
- 2021
- Full Text
- View/download PDF
188. Heregulin-induced cell migration is prevented by trastuzumab and trastuzumab-emtansine in HER2+ breast cancer
- Author
-
Marina Ines Flamini, Joselina Magali Mondaca, Ana Carla Castro Guijarro, and Angel Matias Sanchez
- Subjects
0301 basic medicine ,Scaffold protein ,Cancer Research ,Receptor, ErbB-2 ,Neuregulin-1 ,Breast Neoplasms ,macromolecular substances ,Ado-Trastuzumab Emtansine ,Focal adhesion ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Humans ,Maytansine ,skin and connective tissue diseases ,Paxillin ,Actin nucleation ,biology ,Chemistry ,Actin cytoskeleton reorganization ,Cell migration ,Trastuzumab ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Female ,Signal transduction ,Cortactin - Abstract
Heregulin (HRG) signaling has been implicated in the development of an aggressive phenotype in breast cancer (BC) cells, and HER2 overexpression has been associated with a worse prognosis in BC patients. Nevertheless, the molecular mechanisms through which HRG affects the efficiency of anti-HER2 therapies such as trastuzumab (Tz) and trastuzumab-emtansine (T-DM1) are currently unknown. In the present study, we evaluate the molecular action of HRG toward fundamental scaffold proteins and several kinases in the signal transduction pathways triggered via HER2/HER3, which integrate precise and sequential steps to promote changes in cell morphology to impulse BC cell migration. In addition, we evaluate the effectiveness of Tz and T-DM1 on the control of key proteins involved in BC cell motility, since the acquisition of a migratory phenotype is essential to promote invasion and metastasis. We show that HRG induces actin cytoskeleton reorganization and focal adhesion complex formation, and promotes actin nucleation in BT-474 BC cells. This signaling is triggered by HER2/HER3 to c-Src, FAK and paxillin. When paxillin is phosphorylated, it recruits PAK1, which then phosphorylates cortactin. In parallel, paxillin signals to N-WASP, and both signalings regulate Arp2/3 complex, leading to the local reorganization of actin fibers. Our findings reveal an original mechanism by which HRG increases HER2+ BC cell motility, and show that the latter can be abolished by Tz and T-DM1 treatments. These results provide evidence for the molecular mechanisms involved in cell motility and drug resistance. They will be useful to develop new and more specific therapeutic schemes that interfere with the progression and metastasis of HER2+ BC.
- Published
- 2021
- Full Text
- View/download PDF
189. Simple and Rapid LC–MS/MS Methods for Quantifying Catabolites of Antibody-Drug Conjugates with SMCC Linker
- Author
-
Kelly Dong, Yijue Wu, Fang Chen, Hai-Feng Song, Lihou Dong, Chanrui Wang, and Li Li
- Subjects
Immunoconjugates ,Mass spectrometry ,Mertansine ,Sensitivity and Specificity ,01 natural sciences ,Analytical Chemistry ,Maleimides ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Tandem Mass Spectrometry ,In vivo ,Protein precipitation ,Maytansine ,Chromatography ,010401 analytical chemistry ,Reproducibility of Results ,General Medicine ,0104 chemical sciences ,Standard curve ,chemistry ,030220 oncology & carcinogenesis ,Toxicity ,Linear Models ,Linker ,Chromatography, Liquid ,Conjugate - Abstract
The stability and exposure of toxin-related catabolites in system circulation contributes to the evaluation of the stability, targeted delivery and off-target toxicity for antibody–drug conjugates (ADC) at different stages during drug development. In this study, simple and rapid liquid chromatography–tandem mass spectrometry (LC–MS/MS) methods for determination catabolites of Mertansine (DM1), MCC-DM1 and Lys-MCC-DM1 in cynomolgus serum have been developed. The serum samples are processed by protein precipitation. The LC–MS/MS methods are applied on a Phenomenex C8 column (50 × 2.0 mm, 5 μm) with gradient elution with water–formic acid 0.1% (A) and acetonitrile-formic acid 0.1% (B) at a flow rate of 0.5 mL/min. The analytical run time is only 4.0 min and the calibration ranges of the standard curve are 0.500–200 ng/mL for DM1, 1.00–500 ng/mL for MCC-DM1 and 2.00–1000 ng/mL for Lys-MCC-DM1. Intra- and inter-day precision of low, middle and high quality controls was
- Published
- 2021
- Full Text
- View/download PDF
190. Efflux identification and engineering for ansamitocin P-3 production in Actinosynnema pretiosum
- Author
-
Xinjuan Ning, Xinran Wang, Linquan Bai, Jianhua Wei, Yifan Xiao, and Shuhui Luan
- Subjects
0303 health sciences ,030306 microbiology ,medicine.drug_class ,Mutant ,Antibiotics ,Transporter ,General Medicine ,Biology ,Applied Microbiology and Biotechnology ,Actinobacteria ,Transcriptome ,03 medical and health sciences ,Biochemistry ,Transcription (biology) ,Actinomycetales ,medicine ,Maytansine ,Fermentation ,Efflux ,Gene ,030304 developmental biology ,Biotechnology - Abstract
Ansamitocin P-3 (AP-3) exhibits potent biological activities against various tumor cells. As an important drug precursor, reliable supply of AP-3 is limited by low fermentation yield. Although different strategies have been implemented to improve AP-3 yield, few have investigated the impact of efflux on AP-3 production. In this study, AP-3 efflux genes were identified through combined analysis of two sets of transcriptomes. The production-based transcriptome was implemented to search for efflux genes highly expressed in response to AP-3 accumulation during the fermentation process, while the resistance-based transcriptome was designed to screen for genes actively expressed in response to the exogenous supplementation of AP-3. After comprehensive analysis of two transcriptomes, six efflux genes outside the ansamitocin BGC were identified. Among the six genes, individual deletion of APASM_2704, APASM_6861, APASM_3193, and APASM_2805 resulted in decreased AP-3 production, and alternative overexpression led to AP-3 yield increase from 264.6 to 302.4, 320.4, 330.6, and 320.6 mg/L, respectively. Surprisingly, APASM_2704 was found to be responsible for exportation of AP-3 and another macro-lactam antibiotic pretilactam. Furthermore, growth of APASM_2704, APASM_3193, or APASM_2805 overexpression mutants was obviously improved under 300 mg/L AP-3 supplementation. In summary, our study has identified AP-3 efflux genes outside the ansamitocin BGC by comparative transcriptomic analysis, and has shown that enhancing the transcription of transporter genes can improve AP-3 production, shedding light on strategies used for exporter screening and antibiotic production improvement. KEY POINTS: • AP-3-related efflux genes were identified by transcriptomic analysis. • Deletion of the identified efflux genes led in AP-3 yield decrease. • Overexpression of the efflux genes resulted in increased AP-3 production.
- Published
- 2021
- Full Text
- View/download PDF
191. Therapeutic landscape of advanced HER2-positive breast cancer in 2022
- Author
-
Ruby, Gupta, Sachin, Gupta, Bana, Antonios, Bipin, Ghimire, Vishal, Jindal, Jaskiran, Deol, Suzanna, Gaikazian, Marianne, Huben, Joseph, Anderson, Michael, Stender, and Ishmael, Jaiyesimi
- Subjects
Immunoconjugates ,Receptor, ErbB-2 ,Breast Neoplasms ,Lapatinib ,Trastuzumab ,Ado-Trastuzumab Emtansine ,Phosphatidylinositol 3-Kinases ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Female ,Maytansine ,Taxoids ,Protein Kinase Inhibitors ,Capecitabine - Abstract
HER2-positive breast cancer is an aggressive subtype of breast cancer with five-year survival rates of 30% for the advanced stage. The development of anti-HER2 treatments has led to a paradigm shift in the management and clinical outcomes of advanced HER2-positive breast cancer patients. The standard first-line treatment consists of taxane-based chemotherapy plus dual anti-HER2 therapies with trastuzumab and pertuzumab. The antibody-drug conjugate (ADC) ado-trastuzumab emtansine (T-DM1) has been a second-line therapeutic standard, but the second-line treatment approach is rapidly evolving. Given a substantial advantage of another ADC, Fam-trastuzumab deruxtecan (T-DXd), compared to T-DM1 in a recent randomized trial in the second-line setting, T-DXd is currently the preferred second-line option. Optimal third-line treatment strategies are still not established, and multiple approaches have been used including combinations based on capecitabine, trastuzumab, or both with oral anti-HER2 tyrosine kinase inhibitors. Tucatinib plus capecitabine and trastuzumab, lapatinib plus trastuzumab, neratinib or lapatinib plus capecitabine are some of the FDA approved combinations. Another newer agent approved for third- or later-line therapy in the metastatic setting is margetuximab, an Fc-engineered anti-HER2 monoclonal antibody, in combination with chemotherapy. Other novel agents currently under clinical trials are the drugs that indirectly target HER2, including immune cell cycle inhibitors, PI3K/mTOR inhibitors, and immunotherapy agents.
- Published
- 2022
192. Rational Design and Systemic Appraisal of an EGFR-Targeting Antibody-Drug Conjugate LR-DM1 for Pancreatic Cancer
- Author
-
Mei Zhu, Lei Zhou, Shangjiu Hu, Qingfang Miao, Jianhua Gong, Na Zhang, Guoning Zhang, Minghua Wang, Juxian Wang, Hongwei He, and Yucheng Wang
- Subjects
ErbB Receptors ,Pancreatic Neoplasms ,Immunoconjugates ,Receptor, ErbB-2 ,Cell Line, Tumor ,Drug Discovery ,Molecular Medicine ,Antibodies, Monoclonal ,Humans ,Breast Neoplasms ,Female ,Maytansine ,Trastuzumab - Abstract
By harnessing the payload DM1 and a monoclonal antibody LR004 through a noncleavable linker succinimidyl-4-(
- Published
- 2022
193. Targeted delivery of maytansine to liver cancer cells
- Author
-
Hai-Na, Xie, Yu-Yuan, Chen, Guo-Biao, Zhu, Hai-Hao, Han, Xi-Le, Hu, Zhi-Qiang, Pan, Yi, Zang, Dong-Hao, Xie, Xiao-Peng, He, Jia, Li, and Tony D, James
- Subjects
Cell Line, Tumor ,Liver Neoplasms ,Galactose ,Humans ,Maytansine ,Serum Albumin, Human ,Cell Line - Abstract
A two-dimensional (2D) glycomaterial for targeted delivery of maytansine to liver cancer cells was developed. Host-guest interaction between a galactosyl dye and human serum albumin (HSA) produces supramolecular galactoside-HSA conjugates, which are then used to coat 2D MoS
- Published
- 2022
194. Pinpoint Diagnostic Kit for Heat Stroke by Monitoring Lysosomal pH.
- Author
-
Ying Wen, Weijie Zhang, Tao Liu, Fangjun Huo, and Caixia Yin
- Subjects
- *
HEAT stroke , *LYSOSOMES , *HYDROGEN-ion concentration , *CYTOTOXINS , *MAYTANSINE - Abstract
Heat stroke is one of the most serious causes of mortality. To prevent the situation, it is fundamental to research the mechanism of heat cytotoxicity. The preliminary results revealed that heat stroke and the change of lysosome acidity had some certain correlation. To further clarify their relationship, herein, we report a highly selective and sensitive fluorescence probe (NT1) for turn-on sensing of the pH value. NT2 was synthesized as control compound. Compared to NT2, NT1 showed accurate lysosome target ability. In addition, the suitable pKa value (5.67) allows NT1 to response to the changes of lysosomal pH values. Most importantly, NT1 could be used to study the correlation between the change of lysosomal pH and heat stroke. It was shown that the lysosomal pH value increasing with temperature during heat stroke. Thus, NT1 was an excellent candidate for research of the complex biological mechanism of heat stroke. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
195. Spatial profiling of maytansine during the germination process of Maytenus senegalensis seeds.
- Author
-
Eckelmann, Dennis, Kusari, Souvik, and Spiteller, Michael
- Abstract
The ecological role of maytansine, an important antineoplastic and antimicrobial compound with high cytotoxicity, particularly as a chemical defense compound has remained elusive since its discovery in the 1970s in Maytenus and Putterlickia plants. In the present study, we have used MALDI-imaging-HRMS to visualize the occurrence as well as spatial and temporal distribution of maytansine in a Maytenus senegalensis plant, seeds obtained from the mother plant during seeding stage, through the germination of the seeds, and finally up to the establishment of seedlings (or daughter plants). Although the mother plant was devoid of maytansine, the bioactive compound was found to be distributed in the cotyledons and the endosperm of the seeds with an augmented accretion towards the seed coat. Furthermore, maytansine was always detected in the emerging seedlings, particularly the cortex encompassing the radicle, hypocotyl, and epicotyl. The typical pattern of accumulation of maytansine not only in the seeds but also during germination provides a proof-of-concept that M . senegalensis is ecologically primed to trigger the production of maytansine in vulnerable tissues such as seeds during plant reproduction. By utilizing maytansine as chemical defense compound against predators and/or pathogens, the plant can ensure viability of the seeds and successful germination, thus leading to the next generation of daughter plants. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
196. Antibody-Drug Conjugates (ADCs) for Personalized Treatment of Solid Tumors: A Review.
- Author
-
Lambert, John, Morris, Charles, Lambert, John M, and Morris, Charles Q
- Subjects
AMINOGLYCOSIDES ,ANTIGENS ,ANTINEOPLASTIC agents ,THERAPEUTIC use of immunoglobulins ,THERAPEUTIC use of monoclonal antibodies ,BENZOATES ,OLIGOPEPTIDES ,MOLECULAR structure ,TUMORS ,THERAPEUTICS - Abstract
Attaching a cytotoxic "payload" to an antibody to form an antibody-drug conjugate (ADC) provides a mechanism for selective delivery of the cytotoxic agent to cancer cells via the specific binding of the antibody to cancer-selective cell surface molecules. The first ADC to receive marketing authorization was gemtuzumab ozogamicin, which comprises an anti-CD33 antibody conjugated to a highly potent DNA-targeting antibiotic, calicheamicin, approved in 2000 for treating acute myeloid leukemia. It was withdrawn from the US market in 2010 following an unsuccessful confirmatory trial. The development of two classes of highly potent microtubule-disrupting agents, maytansinoids and auristatins, as payloads for ADCs resulted in approval of brentuximab vedotin in 2011 for treating Hodgkin lymphoma and anaplastic large cell lymphoma, and approval of ado-trastuzumab emtansine in 2013 for treating HER2-positive breast cancer. Their success stimulated much research into the ADC approach, with >60 ADCs currently in clinical evaluation, mostly targeting solid tumors. Five ADCs have advanced into pivotal clinical trials for treating various solid tumors-platinum-resistant ovarian cancer, mesothelioma, triple-negative breast cancer, glioblastoma, and small cell lung cancer. The level of target expression is a key parameter in predicting the likelihood of patient benefit for all these ADCs, as well as for the approved compound, ado-trastuzumab emtansine. The development of a patient selection strategy linked to target expression on the tumor is thus critically important for identifying the population appropriate for receiving treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
197. Targeting HER2 in Breast Cancer: Latest Developments on Treatment Sequencing and the Introduction of Biosimilars
- Author
-
Megan E. Tesch and Karen A. Gelmon
- Subjects
Oncology ,Immunoconjugates ,Pyridines ,Receptor, ErbB-2 ,medicine.medical_treatment ,Ado-Trastuzumab Emtansine ,Medical Oncology ,0302 clinical medicine ,Trastuzumab ,Antineoplastic Combined Chemotherapy Protocols ,Pharmacology (medical) ,Breast ,Molecular Targeted Therapy ,skin and connective tissue diseases ,Oxazoles ,Mastectomy ,Biosimilar ,Neoadjuvant Therapy ,Chemotherapy, Adjuvant ,030220 oncology & carcinogenesis ,Practice Guidelines as Topic ,Neratinib ,Quinolines ,Female ,Pertuzumab ,medicine.drug ,medicine.medical_specialty ,Breast surgery ,Breast Neoplasms ,Antibodies, Monoclonal, Humanized ,Lapatinib ,Disease-Free Survival ,03 medical and health sciences ,Breast cancer ,Internal medicine ,medicine ,Humans ,Maytansine ,Biosimilar Pharmaceuticals ,neoplasms ,business.industry ,medicine.disease ,Clinical trial ,Clinical Trials, Phase III as Topic ,Quinazolines ,Camptothecin ,Neoplasm Recurrence, Local ,business ,030217 neurology & neurosurgery - Abstract
Approximately 20% of all breast cancers overexpress the human epidermal growth factor receptor 2 (HER2). Targeting breast cancer through this vital oncogenic protein has been a major step towards improved patient outcomes. Today, several anti-HER2 agents are in clinical use including: the monoclonal antibodies trastuzumab and pertuzumab; the small molecule inhibitors lapatinib, neratinib, and tucatinib; and the antibody-drug conjugates ado-trastuzumab emtansine and trastuzumab deruxtecan, in some jurisdictions. In addition, several trastuzumab biosimilars have recently been granted regulatory approval in North America and the EU, and are enhancing patient access to HER2-directed therapy. The various agents differ greatly in their side-effect profiles and approved indications, from neoadjuvant and adjuvant use in early disease, to first- and later-line use in metastatic disease. This review discusses the current treatment recommendations for the use of anti-HER2 agents alone and in combination, examines the latest advances in HER2-targeted drugs and how they may be best applied in clinical practice, and provides guidance on optimal sequencing of the growing array of therapeutic options for HER2-positive breast cancer.
- Published
- 2020
- Full Text
- View/download PDF
198. A phase II study of Mirvetuximab Soravtansine in triple-negative breast cancer
- Author
-
Lei Huo, Gaiane M. Rauch, Alyson Clayborn, Tanbin Rahman, Pamela McCarthy, David Ramirez, Elizabeth Ravenberg, Sadia Saleem, Stacy L. Moulder, Clinton Yam, W. Fraser Symmans, Jason B White, James Stec, Sausan Abouharb, Bora Lim, Jennifer K. Litton, Senthil Damodaran, Meghan Sri Karuturi, and Ryan Sun
- Subjects
Adult ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Immunoconjugates ,Disease Response ,Phases of clinical research ,Triple Negative Breast Neoplasms ,Antibodies, Monoclonal, Humanized ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Folate Receptor 1 ,Maytansine ,Pharmacology (medical) ,Prospective Studies ,Adverse effect ,Triple-negative breast cancer ,Pharmacology ,business.industry ,medicine.disease ,Clinical trial ,030104 developmental biology ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Immunohistochemistry ,Female ,business ,Progressive disease - Abstract
Folate receptor alpha (FRα) has been reported to be expressed in up to 80% of triple-negative breast cancers (TNBC) with limited expression in normal tissues, making it a promising therapeutic target. Mirvetuximab soravtansine (mirvetuximab-s) is an antibody drug conjugate which has shown promise in the treatment of FRα-positive solid tumors in early phase clinical trials. Herein, are the results of the first prospective phase II trial evaluating mirvetuximab-s in metastatic TNBC. Patients with advanced, FRα-positive TNBC were enrolled on this study. Mirvetuximab-s was administered at a dose of 6.0 mg/kg every 3 weeks. 96 patients with advanced TNBC consented for screening. FRα staining was performed on tumor tissue obtained from 80 patients. The rate of FRα positivity by immunohistochemistry was 10.0% (8/80). Two patients were treated on study, with best overall responses of stable disease in one and progressive disease in the other. Adverse events were consistent with earlier studies. The study was terminated early due to the low rate of FRα positivity in the screened patient population and lack of disease response in the two patients treated. The observed rate of FRα positivity was considerably lower than previously reported and none of the patients had a partial or complete response. Treatment with mirvetuximab-s should only be further explored in TNBC if an alternate biomarker strategy is developed for patient selection on the basis of additional preclinical data.
- Published
- 2020
- Full Text
- View/download PDF
199. ADVL1522: A phase 2 study of lorvotuzumab mertansine (IMGN901) in children with relapsed or refractory wilms tumor, rhabdomyosarcoma, neuroblastoma, pleuropulmonary blastoma, malignant peripheral nerve sheath tumor, or synovial sarcoma—A Children's Oncology Group study
- Author
-
Donald A. Barkauskas, Mariana M. Cajaiba, Jeffrey S. Dome, Stacey L. Berg, Joseph G. Pressey, James I. Geller, Steve Y. Cho, Brenda J. Weigel, David Hall, Elizabeth Fox, Rachel A. Kudgus, Stephen D. Voss, Malcolm A. Smith, and Joel M. Reid
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Adolescent ,Maximum Tolerated Dose ,Perforation (oil well) ,Malignant peripheral nerve sheath tumor ,Pleuropulmonary blastoma ,Wilms Tumor ,Gastroenterology ,Article ,Lorvotuzumab mertansine ,Neuroblastoma ,Sarcoma, Synovial ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Rhabdomyosarcoma ,medicine ,Humans ,Maytansine ,030212 general & internal medicine ,Child ,business.industry ,Antibodies, Monoclonal ,Wilms' tumor ,medicine.disease ,Survival Analysis ,CD56 Antigen ,Synovial sarcoma ,Treatment Outcome ,Oncology ,Neurofibrosarcoma ,Area Under Curve ,Child, Preschool ,030220 oncology & carcinogenesis ,Female ,business ,Pulmonary Blastoma ,medicine.drug - Abstract
Background Lorvotuzumab mertansine (IMGN901) is an antibody-drug conjugate linking an antimitotic agent (DM1) to an anti-CD56 antibody (lorvotuzumab). Preclinical efficacy has been noted in Wilms tumor, rhabdomyosarcoma, and neuroblastoma. Synovial sarcoma, malignant peripheral nerve sheath tumor (MPNST), and pleuropulmonary blastoma also express CD56. A phase 2 trial of lorvotuzumab mertansine was conducted to assess its efficacy, recommended phase 2 dose, and toxicities. Methods Eligible patients had relapsed after or progressed on standard therapy for their tumor type. Lorvotuzumab mertansine (110 mg/m2 per dose) was administered at the adult recommended phase 2 dose intravenously on days 1 and 8 of 21-day cycles. Dexamethasone premedication was used. Pharmacokinetic samples, peripheral blood CD56-positive cell counts, and tumor CD56 expression were assessed. Results Sixty-two patients enrolled. The median age was 14.3 years (range, 2.8-29.9 years); 35 were male. Diagnoses included Wilms tumor (n = 17), rhabdomyosarcoma (n = 17), neuroblastoma (n = 12), synovial sarcoma (n = 10), MPNST (n = 5), and pleuropulmonary blastoma (n = 1). Five patients experienced 9 dose-limiting toxicities: hyperglycemia (n = 1), colonic fistula (n = 1) with perforation (n = 1), nausea (n = 1) with vomiting (n = 1), increased alanine aminotransferase in cycle 1 (n = 2), and increased alanine aminotransferase in cycle 2 (n = 1) with increased aspartate aminotransferase (n = 1). Non-dose-limiting toxicities (grade 3 or higher) attributed to lorvotuzumab mertansine were rare. The median values of the maximum concentration, half-life, and area under the curve from zero to infinity for DM1 were 0.87 µg/mL, 35 hours, and 27.9 µg/mL h, respectively. Peripheral blood CD56+ leukocytes decreased by 71.9% on day 8. One patient with rhabdomyosarcoma had a partial response, and 1 patient with synovial sarcoma achieved a delayed complete response. Conclusions Lorvotuzumab mertansine (110 mg/m2 ) is tolerated in children at the adult recommended phase 2 dose; clinical activity is limited.
- Published
- 2020
- Full Text
- View/download PDF
200. A Novel HER2-targeted Antibody–drug Conjugate Offers the Possibility of Clinical Dosing at Trastuzumab-equivalent Exposure Levels
- Author
-
Stepan Chuprakov, Fangjiu Zhang, Yun Cheol Kim, David Rabuka, Dominick Yeo, Robyn M. Barfield, Penelope M. Drake, Ayodele O. Ogunkoya, Mark D. Pegram, Maxine Bauzon, and Colin Hickle
- Subjects
musculoskeletal diseases ,0301 basic medicine ,Drug ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,Antibody-drug conjugate ,Immunoconjugates ,Maximum Tolerated Dose ,Receptor, ErbB-2 ,media_common.quotation_subject ,medicine.medical_treatment ,Breast Neoplasms ,Pharmacology ,Ado-Trastuzumab Emtansine ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,In vivo ,Trastuzumab ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Potency ,Maytansine ,Dosing ,skin and connective tissue diseases ,media_common ,Chemotherapy ,business.industry ,Xenograft Model Antitumor Assays ,Rats ,Macaca fascicularis ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Female ,business ,medicine.drug - Abstract
Trastuzumab and the related antibody-drug conjugate (ADC), ado-trastuzumab emtansine (T-DM1), both target HER2-overexpressing cells. Together, these drugs have treatment indications in both early-stage and metastatic settings for HER2+ breast cancer. T-DM1 retains the antibody functionalities of trastuzumab and adds the potency of a cytotoxic maytansine payload. Interestingly, in the clinic, T-DM1 cannot always replace the use of trastuzumab plus chemotherapy administered together as single agents. We hypothesize that this failure may be due in part to the limited systemic exposure achieved by T-DM1 relative to trastuzumab because of toxicity-related dosing constraints on the ADC. We have developed a trastuzumab-based ADC site-specifically conjugated to maytansine through a noncleavable linker. This construct, termed CAT-01-106, has a drug-to-antibody ratio (DAR) of 1.8, approximately half the average DAR of T-DM1, which comprises a mixture of antibodies variously conjugated with DARs ranging from 0-8. The high DAR species present in T-DM1 contribute to its toxicity and limit its clinical dose. CAT-01-106 showed superior in vivo efficacy compared to T-DM1 at equal payload dosing and was equally or better tolerated compared to T-DM1 at equal payload dosing up to 120 mg/kg in Sprague-Dawley rats and 60 mg/kg in cynomolgus monkeys. CAT-01-106 also showed improved pharmacokinetics in rats relative to T-DM1, with 40% higher ADC exposure levels. Together, the data suggest that CAT-01-106 may be sufficiently tolerable to enable clinical dosing at trastuzumab-equivalent exposure levels, combining the functions of both the antibody and the payload in one drug and potentially improving patient outcomes.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.