1
|
Afshari AR, Sanati M, Mollazadeh H, Kesharwani P, Johnston TP, Sahebkar A. Nanoparticle-based drug delivery systems in cancer: A focus on inflammatory pathways. Semin Cancer Biol 2022; 86:860-872. [PMID: 35115226 DOI: 10.1016/j.semcancer.2022.01.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 01/23/2022] [Accepted: 01/23/2022] [Indexed: 12/16/2022]
Abstract
It has become necessary to accept the clinical reality of therapeutic agents targeting the cancer-associated immune system. In recent decades, several investigations have highlighted the role of inflammation in cancer development. It has now been recognized that inflammatory cells secrete mediators, including enzymes, chemokines, and cytokines. These secreted substances produce an inflammatory microenvironment that is critically involved in cancer growth. Inflammation may enhance genomic instability leading to DNA damage, activation of oncogenes, or compromised tumor suppressor activity, all of which may promote various phases of carcinogenesis. Conventional cancer treatment includes surgery, radiation, and chemotherapy. However, treatment failure occurs because current strategies are unable to achieve complete local control due to metastasis. Nanoparticles (NPs) are a broad spectrum of drug carriers typically below the size of 100 nm, targeting tumor sites while reducing off-target consequences. More importantly, NPs can stimulate innate and adaptive immune systems in the tumor microenvironment (TME); hence, they induce a cancer-fighting immune response. Strikingly, targeting cancer cells with NPs helps eliminate drug resistance and tumor recurrence, as well as prevents inflammation. Throughout this review, we provide recent data on the role of inflammation in cancer and explore nano-therapeutic initiatives to target significant mediators, for example, nuclear factor-kappa B (NF-κB), tumor necrosis factor-α (TNF-α), and interleukins (ILs) associated with cancer-related inflammation, to escort the immunomodulators to cancer cells and associated systemic compartments. We also highlight the necessity of better identifying inflammatory pathways in cancer pathophysiology to develop effective treatment plans.
Collapse
Affiliation(s)
- Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Hamid Mollazadeh
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Rebhun RB, York D, Cruz SM, Judge SJ, Razmara AM, Farley LE, Brady RV, Johnson EG, Burton JH, Willcox J, Wittenburg LA, Woolard K, Dunai C, Stewart SL, Sparger EE, Withers SS, Gingrich AA, Skorupski KA, Al-Nadaf S, LeJeune AT, Culp WT, Murphy WJ, Kent MS, Canter RJ. Inhaled recombinant human IL-15 in dogs with naturally occurring pulmonary metastases from osteosarcoma or melanoma: a phase 1 study of clinical activity and correlates of response. J Immunother Cancer 2022; 10:e004493. [PMID: 35680383 PMCID: PMC9174838 DOI: 10.1136/jitc-2022-004493] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2022] [Indexed: 01/04/2023] Open
Abstract
PURPOSE Although recombinant human interleukin-15 (rhIL-15) has generated much excitement as an immunotherapeutic agent for cancer, activity in human clinical trials has been modest to date, in part due to the risks of toxicity with significant dose escalation. Since pulmonary metastases are a major site of distant failure in human and dog cancers, we sought to investigate inhaled rhIL-15 in dogs with naturally occurring lung metastases from osteosarcoma (OSA) or melanoma. We hypothesized a favorable benefit/risk profile given the concentrated delivery to the lungs with decreased systemic exposure. EXPERIMENTAL DESIGN We performed a phase I trial of inhaled rhIL-15 in dogs with gross pulmonary metastases using a traditional 3+3 cohort design. A starting dose of 10 µg twice daily × 14 days was used based on human, non-human primate, and murine studies. Safety, dose-limiting toxicities (DLT), and maximum tolerated dose (MTD) were the primary objectives, while response rates, progression-free and overall survival (OS), and pharmacokinetic and immune correlative analyses were secondary. RESULTS From October 2018 to December 2020, we enrolled 21 dogs with 18 dogs reaching the 28-day response assessment to be evaluable. At dose level 5 (70 μg), we observed two DLTs, thereby establishing 50 µg twice daily × 14 days as the MTD and recommended phase 2 dose. Among 18 evaluable dogs, we observed one complete response >1 year, one partial response with resolution of multiple target lesions, and five stable disease for an overall clinical benefit rate of 39%. Plasma rhIL-15 quantitation revealed detectable and sustained rhIL-15 concentrations between 1-hour and 6 hour postnebulization. Decreased pretreatment lymphocyte counts were significantly associated with clinical benefit. Cytotoxicity assays of banked peripheral blood mononuclear cells revealed significant increases in peak cytotoxicity against canine melanoma and OSA targets that correlated with OS. CONCLUSIONS In this first-in-dog clinical trial of inhaled rhIL-15 in dogs with advanced metastatic disease, we observed promising clinical activity when administered as a monotherapy for only 14 days. These data have significant clinical and biological implications for both dogs and humans with refractory lung metastases and support exploration of combinatorial therapies using inhaled rhIL-15.
Collapse
Affiliation(s)
- Robert B Rebhun
- Department of Surgical and Radiological Sciences, University of California, Davis, California, USA
| | - Daniel York
- Department of Surgical and Radiological Sciences, University of California, Davis, California, USA
| | - Sylvia Margret Cruz
- Division of Surgical Oncology, Department of Surgery, University of California Davis Medical Center, Sacramento, California, USA
| | - Sean J Judge
- Division of Surgical Oncology, Department of Surgery, University of California Davis Medical Center, Sacramento, California, USA
| | - Aryana M Razmara
- Division of Surgical Oncology, Department of Surgery, University of California Davis Medical Center, Sacramento, California, USA
| | - Lauren E Farley
- Division of Surgical Oncology, Department of Surgery, University of California Davis Medical Center, Sacramento, California, USA
| | - Rachel V Brady
- College of Veterinary Medicine, Colorado State University, Fort Collins, Colorado, USA
| | - Eric G Johnson
- Department of Surgical and Radiological Sciences, University of California, Davis, California, USA
| | - Jenna H Burton
- Department of Clinical Sciences, Colorado State University College of Veterinary Medicine, Fort Collins, Colorado, USA
| | - Jennifer Willcox
- Department of Surgical and Radiological Sciences, University of California, Davis, California, USA
| | - Luke A Wittenburg
- Department of Surgical and Radiological Sciences, University of California, Davis, California, USA
| | - Kevin Woolard
- Department of Pathology, University of California, Davis, California, USA
| | - Cordelia Dunai
- Department of Dermatology, University of California, Davis, California, USA
| | - Susan L Stewart
- Department of Public Health Sciences, University of California, Davis, California, USA
| | - Ellen E Sparger
- Department of Medicine and Epidemiology, University of California, Davis, California, USA
| | - Sita S Withers
- Department of Veterinary Clinical Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Alicia A Gingrich
- Division of Surgical Oncology, Department of Surgery, University of California Davis Medical Center, Sacramento, California, USA
| | - Katherine A Skorupski
- Department of Surgical and Radiological Sciences, University of California, Davis, California, USA
| | - Sami Al-Nadaf
- Department of Surgical and Radiological Sciences, University of California, Davis, California, USA
| | - Amandine T LeJeune
- Department of Surgical and Radiological Sciences, University of California, Davis, California, USA
| | - William Tn Culp
- Department of Surgical and Radiological Sciences, University of California, Davis, California, USA
| | - William J Murphy
- Department of Dermatology, University of California Davis Medical Center, Sacramento, California, USA
- Division of Hematology and Oncology, Department of Medicine, University of California Davis Medical Center, Sacramento, California, USA
| | - Michael S Kent
- Department of Surgical and Radiological Sciences, University of California, Davis, California, USA
| | - Robert J Canter
- Division of Surgical Oncology, Department of Surgery, University of California, Davis, California, USA
| |
Collapse
|
3
|
Agarwal Y, Milling LE, Chang JYH, Santollani L, Sheen A, Lutz EA, Tabet A, Stinson J, Ni K, Rodrigues KA, Moyer TJ, Melo MB, Irvine DJ, Wittrup KD. Intratumourally injected alum-tethered cytokines elicit potent and safer local and systemic anticancer immunity. Nat Biomed Eng 2022; 6:129-143. [PMID: 35013574 PMCID: PMC9681025 DOI: 10.1038/s41551-021-00831-9] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 11/05/2021] [Indexed: 02/07/2023]
Abstract
Anti-tumour inflammatory cytokines are highly toxic when administered systemically. Here, in multiple syngeneic mouse models, we show that the intratumoural injection of recombinantly expressed cytokines bound tightly to the common vaccine adjuvant aluminium hydroxide (alum) (via ligand exchange between hydroxyls on the surface of alum and phosphoserine residues tagged to the cytokine by an alum-binding peptide) leads to weeks-long retention of the cytokines in the tumours, with minimal side effects. Specifically, a single dose of alum-tethered interleukin-12 induced substantial interferon-γ-mediated T-cell and natural-killer-cell activities in murine melanoma tumours, increased tumour antigen accumulation in draining lymph nodes and elicited robust tumour-specific T-cell priming. Moreover, intratumoural injection of alum-anchored cytokines enhanced responses to checkpoint blockade, promoting cures in distinct poorly immunogenic syngeneic tumour models and eliciting control over metastases and distant untreated lesions. Intratumoural treatment with alum-anchored cytokines represents a safer and tumour-agnostic strategy to improving local and systemic anticancer immunity.
Collapse
Affiliation(s)
- Yash Agarwal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lauren E Milling
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jason Y H Chang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Luciano Santollani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Allison Sheen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emi A Lutz
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anthony Tabet
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jordan Stinson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kaiyuan Ni
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kristen A Rodrigues
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard-MIT Health Sciences and Technology Program, Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA
| | - Tyson J Moyer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Mariane B Melo
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Darrell J Irvine
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA.
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - K Dane Wittrup
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
4
|
Karimi M, Kamali H, Mohammadi M, Tafaghodi M. Evaluation of various techniques for production of inhalable dry powders for pulmonary delivery of peptide and protein. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
5
|
Zhang KS, Pelleg T, Campbell S, Rubio C, Loschner AL, Ie S. Pulmonary metastatic melanoma: current state of diagnostic imaging and treatments. Melanoma Manag 2021; 8:MMT58. [PMID: 34900220 PMCID: PMC8656320 DOI: 10.2217/mmt-2021-0001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
Melanoma is the deadliest form of skin cancer with an estimated incidence of over 160,000 cases annually and about 41,000 melanoma-related deaths per year worldwide. Malignant melanoma (MM) primarily occurs in the skin but has been described in other organs. Although the respiratory system is generally afflicted by tumors such as lung cancer, it is also rarely affected by primary MM. The estimated incidence of pulmonary MM of the lung accounts for 0.01% of all primary lung tumors. The current understanding of pulmonary MM of the lung pathophysiology and its management are not well established. We aim to survey current clinical modalities with a focus on diagnostic imaging and therapeutic intervention to guide providers in the management of patients with a high index of suspicion.
Collapse
Affiliation(s)
- Kermit S Zhang
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Tomer Pelleg
- Samaritan Health Services, Corvallis, OR 97330, USA
| | - Sabrina Campbell
- Department of Pulmonary & Sleep Medicine, Carilion Clinic, Roanoke, VA 24016, USA
| | - Catalina Rubio
- Department of Basic Science Education, Liberty University College of Osteopathic Medicine, Lynchburg, VA 24502, USA
| | - Anthony Lukas Loschner
- Samaritan Health Services, Corvallis, OR 97330, USA.,Department of Pulmonary & Sleep Medicine, Carilion Clinic, Roanoke, VA 24016, USA
| | - Susanti Ie
- Samaritan Health Services, Corvallis, OR 97330, USA.,Department of Pulmonary & Sleep Medicine, Carilion Clinic, Roanoke, VA 24016, USA
| |
Collapse
|
6
|
Ponkshe P, Feng S, Tan C. Inhalable liposomes for treating lung diseases: clinical development and challenges. Biomed Mater 2021; 16. [PMID: 34134097 DOI: 10.1088/1748-605x/ac0c0c] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 06/16/2021] [Indexed: 12/15/2022]
Abstract
Inhalation delivery of liposomal drugs has distinct advantages for the treatment of pulmonary diseases. Inhalable liposomes of several drugs are currently undergoing clinical trials for a range of indications in the lungs. Herein, general principles of pulmonary delivery as well as the clinical development of inhalable liposomal drugs are reviewed.
Collapse
Affiliation(s)
- Pranav Ponkshe
- Department of Pharmaceutics and Drug Delivery, University of Mississippi School of Pharmacy, University, Mississippi 38655, The United States
| | - Sheng Feng
- Department of Pharmaceutics and Drug Delivery, University of Mississippi School of Pharmacy, University, Mississippi 38655, The United States
| | - Chalet Tan
- Department of Pharmaceutics and Drug Delivery, University of Mississippi School of Pharmacy, University, Mississippi 38655, The United States
| |
Collapse
|
7
|
Donkor M, Jones HP. The Proposition of the Pulmonary Route as an Attractive Drug Delivery Approach of Nano-Based Immune Therapies and Cancer Vaccines to Treat Lung Tumors. FRONTIERS IN NANOTECHNOLOGY 2021. [DOI: 10.3389/fnano.2021.635194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the leading cause of cancer related deaths globally, making it a major health concern. The lung’s permissive rich microenvironment is ideal for supporting outgrowth of disseminated tumors from pre-existing extra-pulmonary malignancies usually resulting in high mortality. Tumors occurring in the lungs are difficult to treat, necessitating the need for the development of advanced treatment modalities against primary tumors and secondary lung metastasis. In this review, we explore the pulmonary route as an attractive drug delivery approach to treat lung tumors. We also discuss the potential of pulmonary delivery of cancer vaccine vectors to induce mucosal immunity capable of preventing the seeding of tumors in the lung.
Collapse
|
8
|
Alhudaithi SS, Almuqbil RM, Zhang H, Bielski ER, Du W, Sunbul FS, Bos PD, da Rocha SRP. Local Targeting of Lung-Tumor-Associated Macrophages with Pulmonary Delivery of a CSF-1R Inhibitor for the Treatment of Breast Cancer Lung Metastases. Mol Pharm 2020; 17:4691-4703. [PMID: 33170724 DOI: 10.1021/acs.molpharmaceut.0c00983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The lungs are major sites of metastases for several cancer types, including breast cancer (BC). Prognosis and quality of life of BC patients that develop pulmonary metastases are negatively impacted. The development of strategies to slow the growth and relieve the symptoms of BC lung metastases (BCLM) is thus an important goal in the management of BC. However, systemically administered first line small molecule chemotherapeutics have poor pharmacokinetic profiles and biodistribution to the lungs and significant off-target toxicity, severely compromising their effectiveness. In this work, we propose the local delivery of add-on immunotherapy to the lungs to support first line chemotherapy treatment of advanced BC. In a syngeneic murine model of BCLM, we show that local pulmonary administration (p.a.) of PLX-3397 (PLX), a colony-stimulating factor 1 receptor inhibitor (CSF-1Ri), is capable of overcoming physiological barriers of the lung epithelium, penetrating the tumor microenvironment (TME), and decreasing phosphorylation of CSF-1 receptors, as shown by the Western blot of lung tumor nodules. That inhibition is accompanied by an overall decrease in the abundance of protumorigenic (M2-like) macrophages in the TME, with a concomitant increase in the amount of antitumor (M1-like) macrophages when compared to the vehicle-treated control. These effects with PLX (p.a.) were achieved using a much smaller dose (1 mg/kg, every other day) compared to the systemic doses typically used in preclinical studies (40-800 mg/kg/day). As an additive in combination with intravenous (i.v.) administration of paclitaxel (PTX), PLX (p.a.) leads to a decrease in tumor burden without additional toxicity. These results suggested that the proposed immunochemotherapy, with regional pulmonary delivery of PLX along with the i.v. standard of care chemotherapy, may lead to new opportunities to improve treatment, quality of life, and survival of patients with BCLM.
Collapse
Affiliation(s)
- Sulaiman S Alhudaithi
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Rashed M Almuqbil
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Hanming Zhang
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Elizabeth R Bielski
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Wei Du
- Department of Pathology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Fatemah S Sunbul
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Paula D Bos
- Department of Pathology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States.,VCU Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Sandro R P da Rocha
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States.,VCU Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
9
|
Das SS, Alkahtani S, Bharadwaj P, Ansari MT, ALKahtani MDF, Pang Z, Hasnain MS, Nayak AK, Aminabhavi TM. Molecular insights and novel approaches for targeting tumor metastasis. Int J Pharm 2020; 585:119556. [PMID: 32574684 DOI: 10.1016/j.ijpharm.2020.119556] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/01/2020] [Accepted: 06/14/2020] [Indexed: 12/18/2022]
Abstract
In recent years, due to the effective drug delivery and preciseness of tumor sites or microenvironment, the targeted drug delivery approaches have gained ample attention for tumor metastasis therapy. The conventional treatment approaches for metastasis therapy have reported with immense adverse effects because they exhibited maximum probability of killing the carcinogenic cells along with healthy cells. The tumor vasculature, comprising of vasculogenic impressions and angiogenesis, greatly depends upon the growth and metastasis in the tumors. Therefore, various nanocarriers-based delivery approaches for targeting to tumor vasculature have been attempted as efficient and potential approaches for the treatment of tumor metastasis and the associated lesions. Furthermore, the targeted drug delivery approaches have found to be most apt way to overcome from all the limitations and adverse effects associated with the conventional therapies. In this review, various approaches for efficient targeting of pharmacologically active chemotherapeutics against tumor metastasis with the cohesive objectives of prognosis, tracking and therapy are summarized.
Collapse
Affiliation(s)
- Sabya Sachi Das
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835 215, Jharkhand, India
| | - Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Priyanshu Bharadwaj
- UFR des Sciences de Santé, Université de Bourgogne Franche-Comté, Dijon 21000, France
| | - Mohammed Tahir Ansari
- School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, Semenyih, Kajang, Selangor 43500, Malaysia
| | - Muneera D F ALKahtani
- Biology Department, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 102275, Riyadh 11675, Saudi Arabia
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai 201203, China
| | - Md Saquib Hasnain
- Department of Pharmacy, Shri Venkateshwara University, NH-24, Rajabpur, Gajraula, Amroha 244236, U.P., India.
| | - Amit Kumar Nayak
- Department of Pharmaceutics, Seemanta Institute of Pharmaceutical Sciences, Mayurbhanj 757086, Odisha, India.
| | | |
Collapse
|
10
|
Verco J, Johnston W, Frost M, Baltezor M, Kuehl PJ, Lopez A, Gigliotti A, Belinsky SA, Wolff R, diZerega G. Inhaled Submicron Particle Paclitaxel (NanoPac) Induces Tumor Regression and Immune Cell Infiltration in an Orthotopic Athymic Nude Rat Model of Non-Small Cell Lung Cancer. J Aerosol Med Pulm Drug Deliv 2019; 32:266-277. [PMID: 31347939 PMCID: PMC6781259 DOI: 10.1089/jamp.2018.1517] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: This study evaluated the antineoplastic and immunostimulatory effects of inhaled (IH) submicron particle paclitaxel (NanoPac®) in an orthotopic non-small cell lung cancer rodent model. Methods: Male nude rats were whole body irradiated, intratracheally instilled with Calu-3 cancer cells and divided into six treatment arms (n = 20 each): no treatment (Group 1); intravenous nab-paclitaxel at 5.0 mg/kg once weekly for 3 weeks (Group 2); IH NanoPac at 0.5 or 1.0 mg/kg, once weekly for 4 weeks (Groups 3 and 4), or twice weekly for 4 weeks (Groups 5 and 6). Upon necropsy, left lungs were paraffin embedded, serially sectioned, and stained for histopathological examination. A subset was evaluated by immunohistochemistry (IHC), anti-pan cytokeratin staining AE1/AE3+ tumor cells and CD11b+ staining dendritic cells, natural killer lymphocytes, and macrophage immune cells (n = 2, Group 1; n = 3 each for Groups 2–6). BCL-6 staining identified B lymphocytes (n = 1 in Groups 1, 2, and 6). Results: All animals survived to scheduled necropsy, exhibited no adverse clinical observations due to treatment, and gained weight at the same rate throughout the study. Histopathological evaluation of Group 1 lung samples was consistent with unabated tumor growth. Group 2 exhibited regression in 10% of animals (n = 2/20). IH NanoPac-treated groups exhibited significantly higher tumor regression incidence per group (n = 11–13/20; p < 0.05, χ2). IHC subset analysis revealed tumor-nodule cluster separation, irregular borders between tumor and non-neoplastic tissue, and an increased density of infiltrating CD11b+ cells in Group 2 animals (n = 2/3) and in all IH NanoPac-treated animals reviewed (n = 3/3 per group). A single animal in Group 4 and Group 6 exhibited signs of pathological complete response at necropsy with organizing stroma and immune cells replacing areas presumed to have previously contained adenocarcinoma nodules. Conclusion: Tumor regression and immune cell infiltration were observed in all treatment groups, with an increased incidence noted in animals receiving IH submicron particle paclitaxel treatment.
Collapse
Affiliation(s)
- James Verco
- US Biotest, Inc., San Luis Obispo, California
| | | | - Michael Frost
- Western Diagnostic Services Laboratory, Santa Maria, California
| | | | | | - Anita Lopez
- Lovelace Biomedical, Albuquerque, New Mexico
| | | | | | | | - Gere diZerega
- US Biotest, Inc., San Luis Obispo, California.,NanOlogy, LLC, Fort Worth, Texas
| |
Collapse
|
11
|
Clinical Response Rates From Interleukin-2 Therapy for Metastatic Melanoma Over 30 Years' Experience: A Meta-Analysis of 3312 Patients. J Immunother 2018; 40:21-30. [PMID: 27875387 DOI: 10.1097/cji.0000000000000149] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Interleukin-2 (IL-2), initially used in 1986, can induce clinical regression-complete responses (CR) and partial responses (PR) of metastatic malignant melanoma. IL-2 has been used alone or in combination, and in different dosage schedules, as an immunotherapeutic agent for melanoma treatment. This meta-analysis aimed to document and evaluate the spectrum of reported clinical response rates from the combined experience of almost 30 years of IL-2 clinical usage. Clinical trials using IL-2 for metastatic melanoma therapy that reported: dosage, combinations, study details, definitions and clinical CR, PR, and overall response (OR) rates were included. A meta-analysis was conducted using the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. In total, 34 studies met inclusion criteria, with 41 separate treatment arms. For all IL-2 treatment modalities collectively, the CR rate was 4.0% [95% confidence interval (CI), 2.8-5.3], PR 12.5% (95% CI, 10.1-15.0), and OR 19.7% (95% CI, 15.9-23.5). CR pre-1994 was 2.7% versus 6.1% post-1994. High and intermediate-IL-2 dosage showed no CR difference, while low-dose IL-2 showed a nonstatistical trend toward an increased CR rate. The highest CR rate resulted from IL-2 combined with vaccine at 5.0%. The meta-analysis showed that IL-2 immunotherapy for advanced metastatic melanoma delivered a CR rate of 4% (range, 0-23%) across nearly 30 years of clinical studies, with gradual improvement over time. The significance is that, contrary to popular belief, the data demonstrated that CR rates were similar for intermediate versus high-IL-2 dosing.
Collapse
|
12
|
Inhalable particulate drug delivery systems for lung cancer therapy: Nanoparticles, microparticles, nanocomposites and nanoaggregates. J Control Release 2018; 269:374-392. [DOI: 10.1016/j.jconrel.2017.11.036] [Citation(s) in RCA: 230] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/21/2017] [Accepted: 11/21/2017] [Indexed: 12/20/2022]
|
13
|
Delivering safer immunotherapies for cancer. Adv Drug Deliv Rev 2017; 114:79-101. [PMID: 28545888 DOI: 10.1016/j.addr.2017.05.011] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 05/05/2017] [Accepted: 05/17/2017] [Indexed: 12/14/2022]
Abstract
Cancer immunotherapy is now a powerful clinical reality, with a steady progression of new drug approvals and a massive pipeline of additional treatments in clinical and preclinical development. However, modulation of the immune system can be a double-edged sword: Drugs that activate immune effectors are prone to serious non-specific systemic inflammation and autoimmune side effects. Drug delivery technologies have an important role to play in harnessing the power of immune therapeutics while avoiding on-target/off-tumor toxicities. Here we review mechanisms of toxicity for clinically-relevant immunotherapeutics, and discuss approaches based in drug delivery technology to enhance the safety and potency of these treatments. These include strategies to merge drug delivery with adoptive cellular therapies, targeting immunotherapies to tumors or select immune cells, and localizing therapeutics intratumorally. Rational design employing lessons learned from the drug delivery and nanomedicine fields has the potential to facilitate immunotherapy reaching its full potential.
Collapse
|
14
|
Morales JO, Fathe KR, Brunaugh A, Ferrati S, Li S, Montenegro-Nicolini M, Mousavikhamene Z, McConville JT, Prausnitz MR, Smyth HDC. Challenges and Future Prospects for the Delivery of Biologics: Oral Mucosal, Pulmonary, and Transdermal Routes. AAPS JOURNAL 2017; 19:652-668. [DOI: 10.1208/s12248-017-0054-z] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 02/01/2017] [Indexed: 12/25/2022]
|
15
|
Abstract
Micrometastatic disease following pulmonary metastasectomy is an ideal setting to test adjuvant immunotherapy, as the efficacy of immunotherapy in experimental models is greatest with the smallest tumor burdens. Although there is not a standard-of-care adjuvant immunotherapy for resected pulmonary metastases, there have been several studies using cytokines and other immunostimulatory molecules in conjunction with metastasectomies in patients with melanoma, renal cell carcinoma, sarcoma, and colorectal cancer, which have provided preliminary data that such adjuvant therapy is feasible and safe and may be useful in the future, following more rigorous testing, as routine therapy to prevent recurrences.
Collapse
Affiliation(s)
- Michael A Morse
- Division of Medical Oncology, Duke University Medical Center, MSRB Room 403, Box 3233, Research Drive, Durham, NC 27710, USA.
| |
Collapse
|
16
|
Ulivieri A, Cardillo G, Manente L, Paone G, Mancuso AP, Vigna L, Di Stasio E, Gasbarra R, Girlando S, Leone A. Molecular characterization of a selected cohort of patients affected by pulmonary metastases of malignant melanoma: Hints from BRAF, NRAS and EGFR evaluation. Oncotarget 2016; 6:19868-79. [PMID: 26305188 PMCID: PMC4637326 DOI: 10.18632/oncotarget.4503] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/20/2015] [Indexed: 02/06/2023] Open
Abstract
Background Melanoma is highly curable in early stages but holds devastating consequences in advanced phases with a median survival of 6–10 months. Lungs are a common metastasis target, but despite this, limited data are available on the molecular status of pulmonary lesions. Materials and Methods 25 patients with surgically resected melanoma lung metastases were screened for BRAF, NRAS, CKIT and EGFR alterations. The results were correlated with time to lung metastasis (TLM), relapse-free survival after metastasectomy (RFS) and overall survival (OS). Results BRAF or NRAS were mutated in 52% and 20% of cases while CKIT was unaffected. Chromosome 7 polysomy was detected in 47% of cases with 17.5% showing EGFR amplification and concomitant BRAF mutation. NRAS mutated patients developed LM within 5 yrs from primary melanoma with larger lesions compared with BRAF (mean diameter 3.3 ± 2.2cm vs 1.9 ± 1.1cm, p = 0.2). NRAS was also associated with a shorter median RFS and OS after metastasectomy. Moreover, Cox regression analysis revealed that NRAS mutation was the only predictive factor of shorter survival from primary melanoma (p = 0.039, OR = 5.5 (1.1–27.6)). Conclusions Molecular characterization identifies advanced melanoma subgroups with distinct prognosis and therapeutic options. The presence of NRAS mutation was associated to a worse disease evolution.
Collapse
Affiliation(s)
- Alessandra Ulivieri
- Anatomic Pathology Unit, San Camillo-Forlanini Hospitals, Rome, Italy.,Laboratory of Biomedical research "Fondazione Niccolò Cusano per la Ricerca Medico-Scientifica" Niccolò Cusano University of Rome, Rome, Italy
| | | | - Liborio Manente
- Anatomic Pathology Unit, San Camillo-Forlanini Hospitals, Rome, Italy
| | - Gregorino Paone
- Department of Respiratory Diseases, San Camillo-Forlanini Hospitals, Rome, Italy
| | | | - Leonardo Vigna
- Department of Medical Oncology, San Camillo-Forlanini Hospitals, Rome, Italy
| | - Enrico Di Stasio
- Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rita Gasbarra
- Anatomic Pathology Unit, San Camillo-Forlanini Hospitals, Rome, Italy
| | | | - Alvaro Leone
- Anatomic Pathology Unit, San Camillo-Forlanini Hospitals, Rome, Italy
| |
Collapse
|
17
|
Co-delivery of chemotherapeutics and proteins for synergistic therapy. Adv Drug Deliv Rev 2016; 98:64-76. [PMID: 26546464 DOI: 10.1016/j.addr.2015.10.021] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 01/15/2023]
Abstract
Combination therapy with chemotherapeutics and protein therapeutics, typically cytokines and antibodies, has been a type of crucial approaches for synergistic cancer treatment. However, conventional approaches by simultaneous administration of free chemotherapeutic drugs and proteins lead to limitations for further optimizing the synergistic effects, due to the distinct in vivo pharmacokinetics and distribution of small drugs and proteins, insufficient tumor selectivity and tumor accumulation, unpredictable drug/protein ratios at tumor sites, short half-lives, and serious systemic adverse effects. Consequently, to obtain optimal synergistic anti-tumor efficacy, considerable efforts have been devoted to develop the co-delivery systems for co-incorporating chemotherapeutics and proteins into a single carrier system and subsequently releasing the dual or multiple payloads at desired target sites in a more controllable manner. The co-delivery systems result in markedly enhanced blood stability and in vivo half-lives of the small drugs and proteins, elevated tumor accumulation, as well as the capability of delivering the multiple agents to the same target sites with rational drug/protein ratios, which may facilitate maximizing the synergistic effects and therefore lead to optimal antitumor efficacy. This review emphasizes the recent advances in the co-delivery systems for chemotherapeutics and proteins, typically cytokines and antibodies, for systemic or localized synergistic cancer treatment. Moreover, the proposed mechanisms responsible for the synergy of chemotherapeutic drugs and proteins are discussed.
Collapse
|
18
|
Rudokas M, Najlah M, Alhnan MA, Elhissi A. Liposome Delivery Systems for Inhalation: A Critical Review Highlighting Formulation Issues and Anticancer Applications. Med Princ Pract 2016; 25 Suppl 2:60-72. [PMID: 26938856 PMCID: PMC5588529 DOI: 10.1159/000445116] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 03/01/2016] [Indexed: 12/18/2022] Open
Abstract
This is a critical review on research conducted in the field of pulmonary delivery of liposomes. Issues relating to the mechanism of nebulisation and liposome composition were appraised and correlated with literature reports of liposome formulations used in clinical trials to understand the role of liposome size and composition on therapeutic outcome. A major highlight was liposome inhalation for the treatment of lung cancers. Many in vivo studies that explored the potential of liposomes as anticancer carrier systems were evaluated, including animal studies and clinical trials. Liposomes can entrap anticancer drugs and localise their action in the lung following pulmonary delivery. The safety of inhaled liposomes incorporating anticancer drugs depends on the anticancer agent used and the amount of drug delivered to the target cancer in the lung. The difficulty of efficient targeting of liposomal anticancer aerosols to the cancerous tissues within the lung may result in low doses reaching the target site. Overall, following the success of liposomes as inhalable carriers in the treatment of lung infections, it is expected that more focus from research and development will be given to designing inhalable liposome carriers for the treatment of other lung diseases, including pulmonary cancers. The successful development of anticancer liposomes for inhalation may depend on the future development of effective aerosolisation devices and better targeted liposomes to maximise the benefit of therapy and reduce the potential for local and systemic adverse effects.
Collapse
Affiliation(s)
- Mindaugas Rudokas
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston
| | - Mohammad Najlah
- Faculty of Medical Science, Anglia Ruskin University, Chelmsford, UK
| | - Mohamed Albed Alhnan
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston
| | - Abdelbary Elhissi
- Pharmaceutical Sciences Section, College of Pharmacy, Qatar University, Doha, Qatar
- *Dr. Abdelbary Elhissi, Pharmaceutical Sciences Section, College of Pharmacy, Qatar University, PO Box 2713, Doha (Qatar), E-Mail
| |
Collapse
|
19
|
Ganapathy V, Moghe PV, Roth CM. Targeting tumor metastases: Drug delivery mechanisms and technologies. J Control Release 2015; 219:215-223. [PMID: 26409123 PMCID: PMC4745901 DOI: 10.1016/j.jconrel.2015.09.042] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/22/2015] [Accepted: 09/22/2015] [Indexed: 12/17/2022]
Abstract
Primary sites of tumor are the focal triggers of cancers, yet it is the subsequent metastasis events that cause the majority of the morbidity and mortality. Metastatic tumor cells exhibit a phenotype that differs from that of the parent cells, as they represent a resistant, invasive subpopulation of the original tumor, may have acquired additional genetic or epigenetic alterations under exposure to prior chemotherapeutic or radiotherapeutic treatments, and reside in a microenvironment differing from that of its origin. This combination of resistant phenotype and distal location make tracking and treating metastases particularly challenging. In this review, we highlight some of the unique biological traits of metastasis, which in turn, inspire emerging strategies for targeted imaging of metastasized tumors and metastasis-directed delivery of therapeutics.
Collapse
Affiliation(s)
- Vidya Ganapathy
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, USA
| | - Prabhas V Moghe
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, USA; Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, USA
| | - Charles M Roth
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, USA; Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, USA.
| |
Collapse
|
20
|
Moraga I, Richter D, Wilmes S, Winkelmann H, Jude K, Thomas C, Suhoski MM, Engleman EG, Piehler J, Garcia KC. Instructive roles for cytokine-receptor binding parameters in determining signaling and functional potency. Sci Signal 2015; 8:ra114. [PMID: 26554818 DOI: 10.1126/scisignal.aab2677] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cytokines dimerize cell surface receptors to activate signaling and regulate many facets of the immune response. Many cytokines have pleiotropic effects, inducing a spectrum of redundant and distinct effects on different cell types. This pleiotropy has hampered cytokine-based therapies, and the high doses required for treatment often lead to off-target effects, highlighting the need for a more detailed understanding of the parameters controlling cytokine-induced signaling and bioactivities. Using the prototypical cytokine interleukin-13 (IL-13), we explored the interrelationships between receptor binding and a wide range of downstream cellular responses. We applied structure-based engineering to generate IL-13 variants that covered a spectrum of binding strengths for the receptor subunit IL-13Rα1. Engineered IL-13 variants representing a broad range of affinities for the receptor exhibited similar potencies in stimulating the phosphorylation of STAT6 (signal transducer and activator of transcription 6). Delays in the phosphorylation and nuclear translocation of STAT6 were only apparent for those IL-13 variants with markedly reduced affinities for the receptor. From these data, we developed a mechanistic model that quantitatively reproduced the kinetics of STAT6 phosphorylation for the entire spectrum of binding affinities. Receptor endocytosis played a key role in modulating STAT6 activation, whereas the lifetime of receptor-ligand complexes at the plasma membrane determined the potency of the variant for inducing more distal responses. This complex interrelationship between extracellular ligand binding and receptor function provides the foundation for new mechanism-based strategies that determine the optimal cytokine dose to enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Ignacio Moraga
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305-5345, USA. Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA
| | - David Richter
- Department of Biology, University of Osnabrück, 49076 Osnabrück, Germany
| | - Stephan Wilmes
- Department of Biology, University of Osnabrück, 49076 Osnabrück, Germany
| | - Hauke Winkelmann
- Department of Biology, University of Osnabrück, 49076 Osnabrück, Germany
| | - Kevin Jude
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305-5345, USA. Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA
| | - Christoph Thomas
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305-5345, USA. Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA
| | - Megan M Suhoski
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA
| | - Edgar G Engleman
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA
| | - Jacob Piehler
- Department of Biology, University of Osnabrück, 49076 Osnabrück, Germany.
| | - K Christopher Garcia
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305-5345, USA. Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA.
| |
Collapse
|
21
|
Kuzmov A, Minko T. Nanotechnology approaches for inhalation treatment of lung diseases. J Control Release 2015; 219:500-518. [PMID: 26297206 DOI: 10.1016/j.jconrel.2015.07.024] [Citation(s) in RCA: 216] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 07/16/2015] [Accepted: 07/21/2015] [Indexed: 02/08/2023]
Abstract
Local administration of therapeutics by inhalation for treatment of lung diseases has the ability to deliver drugs, nucleic acids and peptides specifically to the site of their action and therefore enhance the efficacy of the treatment, limit the penetration of nebulized therapeutic agent(s) into the bloodstream and consequently decrease adverse systemic side effects of the treatment. Nanotechnology allows for a further enhancement of the treatment efficiency. The present review analyzes modern therapeutic approaches of inhaled nanoscale-based pharmaceutics for the detection and treatment of various lung diseases.
Collapse
Affiliation(s)
- Andriy Kuzmov
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway 08854, USA
| | - Tamara Minko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway 08854, USA; Rutgers Cancer Institute of New Jersey, New Brunswick 08903, USA.
| |
Collapse
|
22
|
Efficacy of aerosol therapy of lung cancer correlates with EGFR paralysis induced by AvidinOX-anchored biotinylated Cetuximab. Oncotarget 2015; 5:9239-55. [PMID: 25238453 PMCID: PMC4253431 DOI: 10.18632/oncotarget.2409] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Lung cancer, as well as lung metastases from distal primary tumors, could benefit from aerosol treatment. Unfortunately, because of lung physiology, clearance of nebulized drugs is fast, paralleled by unwanted systemic exposure. Here we report that nebulized AvidinOX can act as an artificial receptor for biotinylated drugs. In nude and SCID mice with advanced human KRAS-mutated A549 metastatic lung cancer, pre-nebulization with AvidinOX enables biotinylated Cetuximab to control tumor growth at a dose lower than 1/25,000 the intravenous effective dose. This result correlates with a striking, specific and unpredictable effect of AvidinOX-anchored biotinylated Cetuximab, as well as Panitumumab, observed on a panel of tumor cell lines, leading to inhibition of dimerization and signalling, blockade of endocytosis, induction of massive lysosomal degradation and abrogation of nuclear translocation of EGFR. Excellent tolerability, together with availability of pharmaceutical-grade AvidinOX and antibodies, will allow rapid clinical translation of the proposed therapy.
Collapse
|
23
|
Zhou Q(T, Tang P, Leung SSY, Chan JGY, Chan HK. Emerging inhalation aerosol devices and strategies: where are we headed? Adv Drug Deliv Rev 2014; 75:3-17. [PMID: 24732364 DOI: 10.1016/j.addr.2014.03.006] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 03/23/2014] [Accepted: 03/24/2014] [Indexed: 02/07/2023]
Abstract
Novel inhaled therapeutics including antibiotics, vaccines and anti-hypertensives, have led to innovations in designing suitable delivery systems. These emerging design technologies are in urgent demand to ensure high aerosolisation performance, consistent efficacy and satisfactory patient adherence. Recent vibrating-mesh and software technologies have resulted in nebulisers that have remarkably accurate dosing and portability. Alternatively, dry powder inhalers (DPIs) have become highly favourable for delivering high-dose and single-dose drugs with the aid of advanced particle engineering. In contrast, innovations are needed to overcome the technical constrains in drug-propellant incompatibility and delivering high-dose drugs with pressurised metered dose inhalers (pMDIs). This review discusses recent and emerging trends in pulmonary drug delivery systems.
Collapse
|