1
|
Maryam S, Krukiewicz K, Haq IU, Khan AA, Yahya G, Cavalu S. Interleukins (Cytokines) as Biomarkers in Colorectal Cancer: Progression, Detection, and Monitoring. J Clin Med 2023; 12:jcm12093127. [PMID: 37176567 PMCID: PMC10179696 DOI: 10.3390/jcm12093127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Cancer is the primary cause of death in economically developed countries and the second leading cause in developing countries. Colorectal cancer (CRC) is the third most common cause of cancer-related deaths worldwide. Risk factors for CRC include obesity, a diet low in fruits and vegetables, physical inactivity, and smoking. CRC has a poor prognosis, and there is a critical need for new diagnostic and prognostic biomarkers to reduce related deaths. Recently, studies have focused more on molecular testing to guide targeted treatments for CRC patients. The most crucial feature of activated immune cells is the production and release of growth factors and cytokines that modulate the inflammatory conditions in tumor tissues. The cytokine network is valuable for the prognosis and pathogenesis of colorectal cancer as they can aid in the cost-effective and non-invasive detection of cancer. A large number of interleukins (IL) released by the immune system at various stages of CRC can act as "biomarkers". They play diverse functions in colorectal cancer, and include IL-4, IL-6, IL-8, IL-11, IL-17A, IL-22, IL-23, IL-33, TNF, TGF-β, and vascular endothelial growth factor (VEGF), which are pro-tumorigenic genes. However, there are an inadequate number of studies in this area considering its correlation with cytokine profiles that are clinically useful in diagnosing cancer. A better understanding of cytokine levels to establish diagnostic pathways entails an understanding of cytokine interactions and the regulation of their various biochemical signaling pathways in healthy individuals. This review provides a comprehensive summary of some interleukins as immunological biomarkers of CRC.
Collapse
Affiliation(s)
- Sajida Maryam
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 44000, Pakistan
| | - Katarzyna Krukiewicz
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland
- Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Konarskiego 22B, 44-100 Gliwice, Poland
| | - Ihtisham Ul Haq
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 44000, Pakistan
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland
- Joint Doctoral School, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
| | - Awal Ayaz Khan
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 44000, Pakistan
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Al Sharqia, Egypt
- Department of Molecular Genetics, Faculty of Biology, Technical University of Kaiserslautern, Paul-Ehrlich Str. 24, 67663 Kaiserslautern, Germany
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| |
Collapse
|
2
|
Yang Z, Loy J, Poirson B, Dai Y, Rajendran S, Xu S, Spires V, Gururajan M, Lin Z, Arbanas J, Carl S, Pace S, Wang Y, Mehl J, Vasudevan K, Spires T, Novosiadly R, Coker S, Perez R, Covello K, Morin P, Graziano R, Broz M, Lehman-McKeeman L. Application of Pharmacokinetic/Pharmacodynamic Modeling to Bridge Mouse Antitumor Efficacy and Monkey Toxicology Data for Determining the Therapeutic Index of an Interleukin-10 Fc Fusion Protein. Front Pharmacol 2022; 13:829063. [PMID: 35795558 PMCID: PMC9251408 DOI: 10.3389/fphar.2022.829063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
Pharmacokinetic/pharmacodynamic (PK/PD) modeling was performed to quantitatively integrate preclinical pharmacology and toxicology data for determining the therapeutic index (TI) of an interleukin-10 (IL-10) fragment crystallizable (Fc) fusion protein. Mouse Fc fused with mouse IL-10 (mFc-mIL-10) was studied in mice for antitumor efficacy, and the elevation of interleukin-18 (IL-18) was examined as a PD biomarker. The in vivo mFc-mIL-10 EC50 for the IL-18 induction was estimated to be 2.4 nM, similar to the in vitro receptor binding affinity (Kd) of 3.2 nM. The IL-18 induction was further evaluated in cynomolgus monkeys, where the in vivo induction EC50 by a human IL-10 human Fc-fusion protein (hFc-hIL-10) was 0.08 nM vs. 0.3 nM measured as the in vitro Kd. The extent of the IL-18 induction correlated with mouse antitumor efficacy and was used to connect mouse efficacy to that in monkeys. The PD-based efficacious dose projected in monkeys was comparable to the results obtained using a PK-based method in which mouse efficacious exposure was targeted and corrected for affinity differences between the species. Furthermore, PK/PD relationships were developed for anemia and thrombocytopenia in monkeys treated with hFc-hIL-10, with thrombocytopenia predicted to be dose-limiting toxicity. Using quantitative pharmacology and toxicology information obtained through modeling work in the same species, the TI of hFc-hIL-10 in monkeys was determined to be 2.4 (vs. PD-based efficacy) and 1.2–3 (vs. PK-based efficacy), indicating a narrow safety margin. The model-based approaches were proven valuable to the developability assessment of the IL-10 Fc-fusion protein.
Collapse
Affiliation(s)
- Zheng Yang
- Metabolism and Pharmacokinetics, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ, United States
- *Correspondence: Zheng Yang,
| | - James Loy
- Discovery Toxicology, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Cambridge, MA, United States
| | - Brian Poirson
- Discovery Oncology, Bristol Myers Squibb, Princeton, NJ, United States
| | - Yanshan Dai
- Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, NJ, United States
| | - Surendran Rajendran
- Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, NJ, United States
| | - Shihua Xu
- Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, NJ, United States
| | - Vanessa Spires
- Discovery Oncology, Bristol Myers Squibb, Princeton, NJ, United States
| | - Murali Gururajan
- Discovery Oncology, Bristol Myers Squibb, Princeton, NJ, United States
| | - Zheng Lin
- Discovery Biotherapeutics, Bristol Myers Squibb, Princeton, NJ, United States
| | - Jaren Arbanas
- Discovery Biotherapeutics, Bristol Myers Squibb, Princeton, NJ, United States
| | - Stephen Carl
- Discovery Pharmaceutics and Analytical Sciences, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ, United States
| | - Samantha Pace
- Discovery Pharmaceutics and Analytical Sciences, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ, United States
| | - Yun Wang
- Discovery Pharmaceutics and Analytical Sciences, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ, United States
| | - John Mehl
- Discovery Pharmaceutics and Analytical Sciences, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ, United States
| | - Krishna Vasudevan
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Thomas Spires
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Ruslan Novosiadly
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Shodeinde Coker
- Oncology Early Clinical Development, Bristol Myers Squibb, Princeton, NJ, United States
| | - Raymond Perez
- Oncology Early Clinical Development, Bristol Myers Squibb, Princeton, NJ, United States
| | - Kelly Covello
- Oncology Early Clinical Development, Bristol Myers Squibb, Princeton, NJ, United States
| | - Paul Morin
- Discovery Biotherapeutics, Bristol Myers Squibb, Princeton, NJ, United States
| | - Robert Graziano
- Discovery Oncology, Bristol Myers Squibb, Princeton, NJ, United States
| | - Miranda Broz
- Discovery Oncology, Bristol Myers Squibb, Redwood City, CA, United States
| | - Lois Lehman-McKeeman
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ, United States
| |
Collapse
|
3
|
Chamanza R, Naylor SW, Carreira V, Amuzie C, Ma JY, Bradley AE, Blankenship B, McDorman K, Louden C. Normal Anatomy, Histology, and Spontaneous Pathology of the Kidney, and Selected Renal Biomarker Reference Ranges in the Cynomolgus Monkey. Toxicol Pathol 2019; 47:612-633. [DOI: 10.1177/0192623319859263] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
To further our understanding of the nonhuman primate kidney anatomy, histology, and incidences of spontaneous pathology, we retrospectively examined kidneys from a total of 505 control Cynomolgus monkeys ( Macaca fascicularis; 264 male and 241 females) aged 2 to 6 years, from toxicity studies. Kidney weights, urinalysis, and kidney-related clinical biochemistry parameters were also evaluated. Although the functional anatomy of the monkey kidney is relatively similar to that of other laboratory animals and humans, a few differences and species-specific peculiarities exist. Unlike humans, the macaque kidney is unipapillate, with a relatively underdeveloped papilla, scarce long loops of Henle, and a near-equivalent cortical to medullary ratio. The most common spontaneous microscopic findings were interstitial infiltrates or interstitial nephritis and other tubular lesions, but several forms of glomerulopathy that may be interpreted as drug-induced were occasionally observed. Common incidental findings of little pathological significance included: papillary mineralization, epithelial pigment, multinucleate cells, cuboidal metaplasia of the Bowman’s capsule, and urothelial inclusions. Kidney weights, and some clinical chemistry parameters, showed age- and sex-related variations. Taken together, these data will aid the toxicologic pathologist to better evaluate the nonhuman primate kidney and assess the species’ suitability as a model for identifying and characterizing drug-induced injury.
Collapse
Affiliation(s)
- Ronnie Chamanza
- Nonclinical Safety, Janssen Research & Development, Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | | | - Vinicius Carreira
- Janssen Pharmaceutical Companies of Johnson & Johnson, La Jolla, CA, USA
| | - Chidozie Amuzie
- Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA, USA
| | - Jing Ying Ma
- Janssen Pharmaceutical Companies of Johnson & Johnson, La Jolla, CA, USA
| | | | | | | | - Calvert Louden
- Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA, USA
| |
Collapse
|
4
|
Chang WS, Shen TC, Yeh WL, Yu CC, Lin HY, Wu HC, Tsai CW, Bau DT. Contribution of Inflammatory Cytokine Interleukin-18 Genotypes to Renal Cell Carcinoma. Int J Mol Sci 2019; 20:ijms20071563. [PMID: 30925760 PMCID: PMC6479470 DOI: 10.3390/ijms20071563] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 12/21/2022] Open
Abstract
Interleukin-18 (IL-18) is a multi-functional immuno-mediator in the development and progression of many types of infectious and inflammatory diseases. In this study, we evaluated the contribution of IL-18 genotypes to renal cell carcinoma (RCC) in Taiwan via the genotyping of IL-18 -656 (A/C), -607 (A/C), and -137 (G/C). Moreover, we analyzed their interactions with smoking, alcohol drinking, hypertension, and diabetes status. The results showed an association of the AC and CC genotypes of IL-18 -607 with a significant decrease in the risk of RCC compared with the AA genotype (odds ratio (OR) = 0.44 and 0.35, 95% confidence interval (CI) = 0.27⁻0.72 and 0.18⁻0.66, p = 0.0008 and 0.0010, respectively). Furthermore, a significantly lower frequency of the C allele at -607 was observed in the RCC group (35.3% vs. 49.8%; OR = 0.53; 95% CI = 0.35⁻0.71, p = 0.0003). However, IL-18 -656 and -137 did not exhibit a likewise differential distribution of these genotypes between the control and case groups. Stratifying the population according to smoking, alcohol drinking, hypertension, and diabetes status revealed a different distribution of IL-18 -607 genotypes among non-smokers, non-drinkers, and patients without diabetes, but not among smokers, drinkers, or patients with diabetes. These findings suggest that IL-18 -607 genotypes may play a role in the etiology and progression of RCC in Taiwan and may serve as a useful biomarker for early detection.
Collapse
Affiliation(s)
- Wen-Shin Chang
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Te-Chun Shen
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Wei-Lan Yeh
- Institute of New Drug Development, China Medical University, Taichung 40402, Taiwan.
| | - Chien-Chih Yu
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan.
| | - Hui-Yi Lin
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan.
| | - Hsi-Chin Wu
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung 40447, Taiwan.
- School of Medicine, China Medical University, Taichung 40402, Taiwan.
| | - Chia-Wen Tsai
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Da-Tian Bau
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung 40447, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan.
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan.
| |
Collapse
|
5
|
Abstract
Anatomically, the kidneys are paired, bean-shaped (in most mammals), excretory organs that lie in the retroperitoneum. High blood flow to the kidneys, together with high oxygen consumption, makes them more vulnerable to exposure, via the circulation, and subsequent injury related to high concentrations of xenobiotics and chemicals. In preclinical drug development and safety assessment of new investigational drugs, changes in kidney structure and/or function following drug administration in experimental laboratory animals need to be put in context with interspecies differences in kidney functional anatomy, physiology, spontaneous pathologies, and toxicopathological responses to injury. In addition, translation to human relevance to avoid premature drug termination from development is vital. Thus, detection and characterization of kidney toxicity in preclinical species and human relevance will depend on the preclinical safety testing strategy and collective weight-of-evidence approach including new investigational drug mechanism of action (MOA), preclinical and clinical interspecies differences, and MOA relevance to humans. This review describes kidney macroscopic and microscopic functional anatomy, physiology, pathophysiology, toxicology, and drug-induced kidney toxicities in safety risk assessment and drug development.
Collapse
Affiliation(s)
- Zaher A Radi
- 1 Pfizer Worldwide Research and Development, Drug Safety R&D, Cambridge, MA, USA
| |
Collapse
|
6
|
Huang CY, Chang WS, Tsai CW, Hsia TC, Shen TC, Bau DT, Shui HA. Interleukin-18 promoter genotype is associated with the risk of nasopharyngeal carcinoma in Taiwan. Cancer Manag Res 2018; 10:5199-5207. [PMID: 30464617 PMCID: PMC6217138 DOI: 10.2147/cmar.s179367] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background The incidence rate of nasopharyngeal carcinoma (NPC) has been documented to be high in Southeast Asia. Interleukin-18 (IL-18) is a multifunctional cytokine that augments interferon-γ production and acts as an important immunomediator in the development of several types of cancer. Patients and methods This case-control study evaluated the role of IL-18 in NPC at the DNA level by genotyping its promoter polymorphisms at positions -656, -607, and -137 in a Taiwanese population. A total of 176 patients with NPC and age- and gender-matched 352 noncancer controls were included in this study. Results The CC genotype of the IL-18-607 polymorphism was found to be associated with significantly decreased risks of NPC compared to the AA genotype (crude OR =0.50, 95% CI =0.29-0.84, P=0.0093). This significant difference persisted even in the dominant and recessive models. A significantly lower C allele frequency at position -607 was detected in the NPC group(41.8% vs 50.3%; OR =0.77; 95% CI =0.63-1.04, P=0.0089). Regarding IL-18-656 and -137 polymorphisms, there were no differential distributions of their genotypes between the NPC and control groups. After substratification of the subjects according to their smoking, alcohol consumption, and areca chewing status, the genotype distribution of the IL-18-607 polymorphism was found to be different only among nonsmokers between the NPC and control subgroups. Conclusion This study suggests that IL-18 plays an important role in the carcinogenesis of NPC in Taiwan and that the genotype-phenotype correlation of IL-18-607 polymorphism and its contribution to NPC need to be investigated further.
Collapse
Affiliation(s)
- Chung-Yu Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Wen-Shin Chang
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan,
| | - Chia-Wen Tsai
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan,
| | - Te-Chun Hsia
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan,
| | - Te-Chun Shen
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan,
| | - Da-Tian Bau
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan, .,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, .,Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan,
| | - Hao-Ai Shui
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
7
|
Doyle SL, Adamson P, López FJ, Humphries P, Campbell M. Reply to IL-18 is not therapeutic for neovascular age-related macular degeneration. Nat Med 2015; 20:1376-7. [PMID: 25473915 DOI: 10.1038/nm.3741] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Sarah L Doyle
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Peter Adamson
- Ophthiris Discovery Performance Unit, GSK, Stevenage, UK
| | - Francisco J López
- Ophthalmology IPE US, RD Alternative Discovery and Development, GSK, King of Prussia, Pennsylvania, USA
| | - Peter Humphries
- Ocular Genetics Unit, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Matthew Campbell
- Ocular Genetics Unit, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
8
|
Singh PK, Ahmad MK, Kumar V, Hussain SR, Gupta R, Jain A, Mahdi AA, Bogra J, Chandra G. Effects of interleukin-18 promoter (C607A and G137C) gene polymorphisms and their association with oral squamous cell carcinoma (OSCC) in northern India. Tumour Biol 2014; 35:12275-84. [DOI: 10.1007/s13277-014-2538-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 08/20/2014] [Indexed: 12/12/2022] Open
|
9
|
Everds NE, Tarrant JM. Unexpected hematologic effects of biotherapeutics in nonclinical species and in humans. Toxicol Pathol 2013; 41:280-302. [PMID: 23471185 DOI: 10.1177/0192623312467400] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Biotherapeutics are expanding the arsenal of therapeutics available for treating and preventing disease. Although initially thought to have limited side effects due to the specificity of their binding, these drugs have now been shown to have potential for adverse drug reactions including effects on peripheral blood cell counts or function. Hematotoxicity caused by a biotherapeutic can be directly related to the activity of the biotherapeutic or can be indirect and due to autoimmunity, biological cascades, antidrug antibodies, or other immune system responses. Biotherapeutics can cause hematotoxicity primarily as a result of cellular activation, cytotoxicity, drug-dependent and independent immune responses, and sequelae from initiating cytokine and complement cascades. The underlying pathogenesis of biotherapeutic-induced hematotoxicity often is poorly understood. Nonclinical studies have generally predicted clinical hematotoxicity for recombinant cytokines and growth factors. However, most hematologic liabilities of biotherapeutics are not based on drug class but are species specific, immune-mediated, and of low incidence. Despite the potential for unexpected hematologic toxicity, the risk-benefit profile of most biotherapeutics is favorable; hematologic effects are readily monitorable and managed by dose modification, drug withdrawal, and/or therapeutic intervention. This article reviews examples of biotherapeutics that have unexpected hematotoxicity in nonclinical or clinical studies.
Collapse
|
10
|
Everds N, Li N, Bailey K, Fort M, Stevenson R, Jawando R, Salyers K, Jawa V, Narayanan P, Stevens E, He C, Nguyen MP, Tran S, Doyle N, Poitout-Belissent F, Jolette J, Xu C, Sprugel K. Unexpected Thrombocytopenia and Anemia in Cynomolgus Monkeys Induced by a Therapeutic Human Monoclonal Antibody. Toxicol Pathol 2013; 41:951-69. [DOI: 10.1177/0192623312474727] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cynomolgus monkeys dosed with a therapeutic monoclonal antibody (mAbY.1) at ≥50 mg/kg had unexpected acute thrombocytopenia (nadir ∼3,000 platelets/µl), sometimes with decreases in red cell mass. Increased activated macrophages, mitotic figures, and erythrophagocytosis were observed in the spleen. Binding of mAbY.1 to cynomolgus peripheral blood cells could not be detected in vitro. mAbY.1 induced phagocytosis of platelets by peripheral blood monocytes from cynomolgus monkeys, but not from humans. mAbs sharing the same constant domain (Fc) sequences, but differing from mAbY.1 in their variable domains, bound competitively to and had similar biological activity against the intended target. None of these antibodies had hematologic liabilities in vitro or in vivo. Neither the F(ab’)2 portion of mAbY.1 nor the F(ab’)2 portion on an aglycosylated Fc (IgG1) framework caused phagocytosis of platelets in vitro. These data suggest that the hematologic effects of mAbY.1 in cynomolgus monkeys likely occurred through an off-target mechanism, shown to be driven by 1 to 3 amino acid differences in the light chain. The hematologic effects made mAbY.1 an unsuitable candidate for further development as a therapeutic agent. This example demonstrates that nonclinical safety studies may be essential for understanding off-target effects of mAbs prior to clinical trials.
Collapse
Affiliation(s)
| | - Nianyu Li
- Amgen Inc., Seattle, Washington, USA
| | - Keith Bailey
- Oklahoma State University, Stillwater, Oklahoma, USA
| | | | | | | | - Kevin Salyers
- Amgen Inc., Thousand Oaks, California, USA
- Kevin Salyers is deceased
| | - Vibha Jawa
- Amgen Inc., Thousand Oaks, California, USA
| | | | | | - Ching He
- Amgen Inc., Seattle, Washington, USA
| | | | - Sam Tran
- Amgen Inc., Seattle, Washington, USA
| | - Nancy Doyle
- Charles River Preclinical Services Montreal, Senneville, Quebec, Canada
| | | | - Jacquelin Jolette
- Charles River Preclinical Services Montreal, Senneville, Quebec, Canada
| | - Cen Xu
- Amgen Inc., Thousand Oaks, California, USA
| | | |
Collapse
|
11
|
Waggie KS, Holdren MS, Byrnes-Blake K, Pedersen S, Ponce R, Hughes S, Miller DM. Preclinical Safety, Pharmacokinetics, and Pharmacodynamics of Recombinant Human Interleukin-21 in Cynomolgus Macaques (Macaca fascicularis). Int J Toxicol 2012; 31:303-16. [DOI: 10.1177/1091581812449661] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Interleukin-21 (IL-21), a pleiotropic immunostimulatory type I cytokine, has anticancer effects in animal models. Preclinical studies designed to assess the safety of recombinant human IL-21 (rIL-21) for use in phase I oncology studies are described. The rIL-21 (≤3.0 mg/kg per dose) was given intravenously to cynomolgus monkeys ( Macaca fascicularis) once daily for 5 days, followed by 9 nondosing days (1 cycle) for ≤4 cycles. The rIL-21 pharmacokinetics was dose-dependent. Accumulation was not observed after repeated dosing, consistent with the short elimination half-life ( t1/2,λz; 0.4-0.8 hours). Safety findings included cyclical anemia and thrombocytopenia, clinical pathology changes consistent with acute-phase response, leukocyte infiltrates in hepatic sinusoids, and sporadic serum transaminase elevations (typically <3 times upper-limit of normal); all were reversible upon cessation of treatment. Decreased pharmacodynamic responses with time corresponded to the development of anti-rIL-21 antibodies; effects varied among individuals and were dose-dependent. These studies demonstrated rIL-21 to be generally well-tolerated when administered to cynomolgus monkeys, and all adverse effects were reversible upon treatment cessation. However, development of anti-rIL-21 antibodies may limit the use of this species in long-term studies.
Collapse
Affiliation(s)
- Kimberly S. Waggie
- ZymoGenetics, a Bristol-Myers Squibb Company, Preclinical Development, Seattle, WA 98102, USA
| | - Matthew S. Holdren
- ZymoGenetics, a Bristol-Myers Squibb Company, Preclinical Development, Seattle, WA 98102, USA
| | - Kelly Byrnes-Blake
- ZymoGenetics, a Bristol-Myers Squibb Company, Preclinical Development, Seattle, WA 98102, USA
- Northwest PK Solutions, Sultan, WA, USA
| | - Susan Pedersen
- ZymoGenetics, a Bristol-Myers Squibb Company, Preclinical Development, Seattle, WA 98102, USA
- Amgen, Inc, Seattle, WA, USA
| | - Rafael Ponce
- ZymoGenetics, a Bristol-Myers Squibb Company, Preclinical Development, Seattle, WA 98102, USA
- Amgen, Inc, Seattle, WA, USA
| | - Steven Hughes
- ZymoGenetics, a Bristol-Myers Squibb Company, Preclinical Development, Seattle, WA 98102, USA
- Dendreon, Seattle, WA, USA
| | - Dennis M. Miller
- ZymoGenetics, a Bristol-Myers Squibb Company, Preclinical Development, Seattle, WA 98102, USA
- CPTxBio LLC, Woodinville WA, USA
| |
Collapse
|
12
|
Farjadfar A, Mojtahedi Z, Ghayumi MA, Erfani N, Haghshenas MR, Ghaderi A. Interleukin-18 promoter polymorphism is associated with lung cancer: a case-control study. Acta Oncol 2010; 48:971-6. [PMID: 19642044 DOI: 10.1080/02841860902878145] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Interleukin-18 (IL-18) is a multifunctional cytokine that augments IFN-gamma production and affects tumor immune response. In the present case-control study, we tested whether IL-18 promoter polymorphism contributes to lung cancer susceptibility in Iranian patients. MATERIAL AND METHODS The study groups were 73 patients with lung cancer, including 53 with squamous carcinoma (SC) and 20 with small cell lung carcinoma (SCLC), and 97 healthy regional aged-matched individuals. The frequency of IL-18 promoter single nucleotide polymorphisms (SNPs) at positions -656 (G/T), -607 (C/A), and -137 (G/C) was determined by polymerase chain reaction analyses. RESULTS There were significant differences in the IL-18 -607 allele and genotype distributions between the 73 lung cancer patients and controls. A significantly higher A allele frequency at position -607, which is associated with lower IL-18 production, was observed in lung cancer patients (48.6% vs. 35%; OR = 1.75; 95% CI 1.13-2.72). Also, patients with the -607 CA and the AA genotypes had a 2.60-fold (95% CI 1.26-5.36) and 3.15-fold (95% CI 1.16-8.55) increase in risk of lung cancer. Subdivision of the patients according to histological type revealed that SC was significantly associated with IL-18 -607 SNPs. Although the percentages of -607 alleles and genotypes in SCLC patients were similar to the results in SC patients, the differences compared to control individuals did not reach statistical significance. Analysis with Arlequin software identified eight haplotypes from three SNPs analyzed here. The distributions of IL-18 gene haplotypes were not significantly different between patients and controls after Bonferroni correction. DISCUSSION This is the first report to investigate the association between IL-18 polymorphism and lung cancer. Our results suggest that IL-18 polymorphism contributes to the lung cancer risk, particularly among SC patients. Further studies with larger numbers of patients are required to determine the possible association between IL-18 polymorphisms and different histological types of lung cancer.
Collapse
|
13
|
Immunogenicity of biologically-derived therapeutics: assessment and interpretation of nonclinical safety studies. Regul Toxicol Pharmacol 2009; 54:164-82. [PMID: 19345250 DOI: 10.1016/j.yrtph.2009.03.012] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 03/24/2009] [Accepted: 03/25/2009] [Indexed: 11/20/2022]
Abstract
An evaluation of potential antibody formation to biologic therapeutics during the course of nonclinical safety studies and its impact on the toxicity profile is expected under current regulatory guidance and is accepted standard practice. However, approaches for incorporating this information in the interpretation of nonclinical safety studies are not clearly established. Described here are the immunological basis of anti-drug antibody formation to biopharmaceuticals (immunogenicity) in laboratory animals, and approaches for generating and interpreting immunogenicity data from nonclinical safety studies of biotechnology-derived therapeutics to support their progression to clinical evaluation. We subscribe that immunogenicity testing strategies should be adapted to the specific needs of each therapeutic development program, and data generated from such analyses should be integrated with available clinical and anatomic pathology, pharmacokinetic, and pharmacodynamic data to properly interpret nonclinical studies.
Collapse
|
14
|
Guzman RE, Datta K, Khan NK. Obstructive Protein Cast Nephropathy in Cynomolgus Monkeys Treated with Small Organic Molecules. Vet Pathol 2008; 45:945-8. [DOI: 10.1354/vp.45-6-945] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have observed a renal toxicity consistent with an obstructive protein cast nephropathy in cynomolgus macaques but not in other species treated with different therapeutic candidates having a common carboxylic acid moiety, suggesting a species-specific sensitivity. Here, we present renal toxicity findings consistent with a protein cast nephropathy in a 2–week safety study in cynomolgus monkeys. Light microscopic changes consisted of intratubular cast formation, tubular dilatation, interstitial inflammation, and expansion of the medullary interstitium. Tubular cast material was identified as Tamm-Horsfall protein (THP) and, on ultrastructure, crystalloid material was present in vacuoles of tubular epithelium. It is hypothesized that microcrystal formation in the urinary tubular spaces induces aggregation of THP protein and cast formation in monkeys. Drug-induced obstructive nephropathy is not identified as a major problem in humans; thus, the clinical relevance of the above findings in monkeys is not clear.
Collapse
Affiliation(s)
- R. E. Guzman
- Drug Safety Research and Development, Pfizer Global Research and Development, Groton, CT
| | - K. Datta
- Drug Safety Research and Development, Pfizer Global Research and Development, Groton, CT
| | - N. K. Khan
- Drug Safety Research and Development, Pfizer Global Research and Development, Groton, CT
| |
Collapse
|
15
|
Chaudhry UI, Kingham TP, Plitas G, Katz SC, Raab JR, DeMatteo RP. Combined stimulation with interleukin-18 and CpG induces murine natural killer dendritic cells to produce IFN-gamma and inhibit tumor growth. Cancer Res 2006; 66:10497-504. [PMID: 17079471 DOI: 10.1158/0008-5472.can-06-1908] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Natural killer dendritic cells (NKDC) are a novel subtype of dendritic cells with natural killer (NK) cell properties. IFN-gamma is a pleiotropic cytokine that plays an important role in the innate immune response to tumors. Based on our previous finding that the combination of Toll-like receptor 9 ligand CpG and interleukin (IL)-4 stimulates NKDC to produce IFN-gamma, we hypothesized that NKDC are the major IFN-gamma-producing dendritic cell subtype and may play a significant role in the host antitumor response. We found that under several conditions in vitro and in vivo NKDC accounted for the majority of IFN-gamma production by murine spleen CD11c(+) cells. IL-18 alone induced NKDC to secrete IFN-gamma, and the combination of IL-18 and CpG resulted in a synergistic increase in IFN-gamma production, both in vitro and in vivo. NK cells made 26-fold less IFN-gamma under the same conditions in vitro, whereas dendritic cells produced a negligible amount. The mechanism of IFN-gamma secretion by NKDC depended on IL-12. NKDC selectively proliferated in vitro and in vivo in response to the combination of IL-18 and CpG. Systemic treatment with IL-18 and CpG reduced the number of B16F10 melanoma lung metastases. The mechanism depended on NK1.1(+) cells, as their depletion abrogated the effect. IL-18 and CpG activated NKDC provided greater tumor protection than NK cells in IFN-gamma(-/-) mice. Thus, NKDC are the major dendritic cell subtype to produce IFN-gamma. The combined use of IL-18 and CpG is a viable strategy to potentiate the antitumor function of NKDC.
Collapse
Affiliation(s)
- Umer I Chaudhry
- Hepatobiliary Service, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | | | | | | | | | | |
Collapse
|
16
|
Lagarce F, Garcion E, Faisant N, Thomas O, Kanaujia P, Menei P, Benoit JP. Development and characterization of interleukin-18-loaded biodegradable microspheres. Int J Pharm 2006; 314:179-88. [PMID: 16515850 DOI: 10.1016/j.ijpharm.2005.07.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2005] [Accepted: 07/02/2005] [Indexed: 01/14/2023]
Abstract
Immunostimulation represents a promising approach designed to specifically eradicate malignant cells. Since glioma tumour cells hole up in the central nervous system (CNS) in a particularly inauspicious milieu to antitumour immune reactions we here propose a new strategy to revert the properties of this microenvironment by administering an antitumour cytokine into the CNS tumour itself. Thus, biodegradable poly(D,L-lactide-co-glycolide) (PLGA) sustained-release microspheres for stereotaxic implantation loaded with interleukin-18 (IL-18), that is known to exert antitumour activity and trigger immune cell-mediated cytotoxicity, were developed. Different tests for assessing IL-18 bioactivity were set-up and evaluated. A specific bioassay was considered as the most reliable test. The stability and integrity of IL-18 was then verified during the encapsulation process. Consequently, two procedures of IL-18 encapsulation in PLGA microparticles (W/O/W and S/O/W) were investigated. As determined by radiolabelling studies using 125I-IL-18 and a continuous flow system, the in vitro release profile of IL-18 was optimum with S/O/W method with a moderate burst effect and a subsequent progressive discharge of 16.5+/-8.4 ng/day during the next 21 days against 6.1+/-4.2 ng/day with the W/O/W method. Considering analytical testing of IL-18 together with its preserved biological activity after release from microspheres, amounts of the active cytokine obtained with S/O/W method were relevant to plan in vivo evaluation to validate the therapeutic strategy.
Collapse
Affiliation(s)
- F Lagarce
- Inserm, U646, 10 rue André Boquel, F-49100 Angers, France
| | | | | | | | | | | | | |
Collapse
|
17
|
Fischer CP, Perstrup LB, Berntsen A, Eskildsen P, Pedersen BK. Elevated plasma interleukin-18 is a marker of insulin-resistance in type 2 diabetic and non-diabetic humans. Clin Immunol 2005; 117:152-60. [PMID: 16112617 DOI: 10.1016/j.clim.2005.07.008] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2005] [Revised: 07/19/2005] [Accepted: 07/20/2005] [Indexed: 12/22/2022]
Abstract
Elevated plasma IL-18 is present in several conditions sharing insulin-resistance as common trait, but the association with insulin-resistance per se is not established. Plasma/serum IL-6, IL-18, TNF-alpha, soluble TNF receptor II (sTNFR2), and C-reactive protein (CRP) were measured in 97 patients with type 2 diabetes (DM) and 84 non-diabetic controls (CON). The association with insulin-resistance-estimated using the homeostasis model assessment (HOMA-IR)-was analyzed using multivariate linear and logistic regression. Compared to CON, DM demonstrated higher plasma levels of IL-18 (P = 0.001), IL-6 (P < 0.001), sTNFR2 (P = 0.005), and CRP (P < 0.001), while TNF-alpha was lower (P = 0.017). Plasma IL-18 increased across HOMA-IR quartiles in both DM and CON, both with and without adjustment for confounders (all P < 0.05). In contrast, neither IL-6, TNF-alpha, sTNFR2, nor CRP was associated with HOMA-IR in CON when adjusting for confounders. Accordingly, 50% increase of IL-18 corresponded to a marked increase of HOMA-IR in both DM and CON (DM: 26%, P = 0.014; CON: 25%, P = 0.003) after adjustment for confounders. Our results show that plasma IL-18 was associated with HOMA-IR independent of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Christian P Fischer
- Centre of Inflammation and Metabolism, The Department of Infectious Diseases and The Copenhagen Muscle Research Centre, Rigshospitalet and The Faculty of Health Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark.
| | | | | | | | | |
Collapse
|
18
|
Omoya K, Kato Z, Matsukuma E, Li A, Hashimoto K, Yamamoto Y, Ohnishi H, Kondo N. Systematic optimization of active protein expression using GFP as a folding reporter. Protein Expr Purif 2005; 36:327-32. [PMID: 15249057 DOI: 10.1016/j.pep.2004.04.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2004] [Revised: 04/27/2004] [Indexed: 11/27/2022]
Abstract
Many recombinant proteins have been used as drugs; however, human proteins expressed using heterologous hosts are often insoluble. To obtain correctly folded active proteins, many optimizations of expression have been attempted but usually are found to be applicable only for specific targets. Interleukin-18 (IL-18) has a key role in many severe disorders including autoimmune diseases, and therapeutic approaches using IL-18 have been reported. However, production of IL-18 in Escherichia coli resulted in extensive inclusion body formation and previous conventional screenings of expression conditions could obtain only a condition with a low yield. To address the problem, we applied a folding reporter system using green fluorescent protein (GFP) for screening of the expression conditions for hIL-18. The established system efficiently screened many conditions, and optimized conditions for the expression of hIL-18 significantly enhanced the final yield of the active protein. Systematic screening using a GFP reporter system could be applied for the production of other proteins and in other organisms.
Collapse
Affiliation(s)
- Kentaro Omoya
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Yanagido 1-1, Gifu, Gifu 501-1194, Japan
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Yamamoto Y, Kato Z, Matsukuma E, Li A, Omoya K, Hashimoto K, Ohnishi H, Kondo N. Generation of highly stable IL-18 based on a ligand–receptor complex structure. Biochem Biophys Res Commun 2004; 317:181-6. [PMID: 15047165 DOI: 10.1016/j.bbrc.2004.03.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2004] [Indexed: 11/30/2022]
Abstract
Human interleukin-18 (hIL-18), initially cloned as an IFN-gamma-inducing factor, has a key role in many inflammatory diseases. We have previously developed a high production system for correctly folded active hIL-18 protein, leading to the revelation of the 3D-structure and the receptor binding mode. These findings can strongly indicate the experimental and medical applications of IL-18; however, the recombinant protein is prone to be inactivated forming multimers. Recently, therapeutic approaches using recombinant IL-18 have shown the effectiveness for treatment of cancer; indicating the necessity of a more stable protein for therapy with intertrial reliability. Here we have generated a highly stable hIL-18 with replacement of cysteine by serine based on the tertiary structure and the binding mechanism, retaining the biological activity. Similar rational designs can be applied to develop new therapeutic molecules of other cytokines.
Collapse
Affiliation(s)
- Yutaka Yamamoto
- Department of Pediatrics, Gifu University School of Medicine, Tsukasa 40, Gifu 500-8705, Japan
| | | | | | | | | | | | | | | |
Collapse
|