1
|
Barbora A, Rahamim S, Bohnick M, Firer MA, Yahalom A. The non-ionizing electromagnetic stimulation enhanced antibody production (NESEAP) effect - Discovery and technological applications. Biotechnol J 2024; 19:e2300277. [PMID: 37753941 DOI: 10.1002/biot.202300277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/10/2023] [Accepted: 09/26/2023] [Indexed: 09/28/2023]
Abstract
The rise of biological therapeutics in the global pharmaceuticals market has escalated the demand for quality monoclonal antibodies for healthcare and scientific applications. Reducing costs while enhancing production yields without compromising quality are the main challenges to the growth of this industry today. Over the last two decades non-ionizing radiation has been demonstrated to elicit targeted biological responses in a frequency and dose dependent manner. We hypothesize and design a millimeter wave radiation procedure to enhance the yields of antibody-producing hybridoma cell lines. We demonstrate this method enhances the production of IgA and IgG antibodies from MOPC315.BM and U13.6 cells by a factor of 24.05 ± 3.32 and 1.41 ± 0.03 respectively relative to untreated cells. No treatment associated cytotoxicity was observed in either cell line corroborating physiological viability of irradiated cells. Our results demonstrate proof-of-concept of a novel technique to significantly enhance antibody yields from hybridoma cells which could lead to a reduction in antibody production costs. Further studies will focus on scaling up of this technology and employment of non-contact, tuned electromagnetic stimulation of biological systems for targeted responses.
Collapse
Affiliation(s)
- Ayan Barbora
- Department of Physics, Ariel University, Ariel, Israel
| | - Shmuel Rahamim
- Department of Chemical Engineering and Biotechnology, Ariel University, Ariel, Israel
| | - Maya Bohnick
- Department of Chemical Engineering and Biotechnology, Ariel University, Ariel, Israel
| | - Michael A Firer
- Department of Chemical Engineering and Biotechnology, Ariel University, Ariel, Israel
- The Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Asher Yahalom
- Department of Physics, Ariel University, Ariel, Israel
- Department of Electrical and Electronic Engineering, Ariel University, Ariel, Israel
| |
Collapse
|
2
|
Pang J, Meng L, Huang H, Ma J, He L, Huang P. Decorated gold nanoparticles on hydroxymethylated lignin modified magnetic composite: Introducing a novel therapeutic drug for the treatment of renal anemia. INORG CHEM COMMUN 2023. [DOI: 10.1016/j.inoche.2023.110686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
|
3
|
Cai Y, Karmakar B, Babalghith AO, Batiha GES, AlSalem HS, El-Kott AF, Shati AA, Alfaifi MY, Elbehairi SEI. Decorated Au NPs on lignin coated magnetic nanoparticles: Investigation of its catalytic application in the reduction of aromatic nitro compounds and its performance against human lung cancer. Int J Biol Macromol 2022; 223:1067-1082. [PMID: 36368366 DOI: 10.1016/j.ijbiomac.2022.10.268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 10/17/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
Abstract
In the recent years, bio-functionalized noble metal doped advanced magnetics nanocomposite materials has been materialized as potential featured catalysts in diverse applications. In this connection, we report herein a novel biogenic lignin driven Au nanoparticle supported Fe3O4 composite material. The procedure is free from any harsh reducing or stabilizing agent. Morphology and structural features were assessed following different physicochemical methodologies like FT-IR, FE-SEM, TEM, EDS, XRD, VSM and ICP-OES techniques. Thereafter, the [Fe3O4/Lignin/Au] material was successfully employed in the efficient reduction of different nitroarenes in aqueous medium. The process was monitored over UV-Vis spectroscopic study. Excellent yields were achieved with a range of diverse functionalized nitroarenes within 10-45 min of reaction. The nanocatalyst was recycled 10 times without any significant loss of catalytic activity. Distinctiveness of the material's activity was validated by comparing the results in the reduction of 4-nitrophenol. Furthermore, the prepared [Fe3O4/Lignin/Au] nanocomposite system exhibited outstanding antioxidant and anticancer effects against five lung cancer cell lines, such as, BICR 3, BICR 78, CALU 1, ChaGo-K-1, and A549. Cytotoxicity assay was determined in terms of % cell viability following MTT protocol. The corresponding IC50 values were obtained as 47, 31, 19, 25, and 31 μg/mL respectively.
Collapse
Affiliation(s)
- Yi Cai
- Department of Medical Oncology, Chinese PLA General Hospital & Medical School, Beijing 100853, China
| | - Bikash Karmakar
- Department of Chemistry, Gobardanga Hindu College, 24 Parganas (North), India
| | - Ahmad O Babalghith
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
| | - Huda S AlSalem
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Attalla F El-Kott
- Department of Biology, Faculty of Science, King Khalid University, Abha, Saudi Arabia; Department of Zoology, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Ali A Shati
- Department of Biology, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Y Alfaifi
- Department of Biology, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Serag Eldin I Elbehairi
- Department of Biology, Faculty of Science, King Khalid University, Abha, Saudi Arabia; Cell Culture Lab, Egyptian Organization for Biological Products and Vaccines (VACSERA Holding Company), 51 Wezaret El-Zeraa St., Agouza, Giza, Egypt.
| |
Collapse
|
4
|
Gold Nanoparticles in Diagnostics and Therapeutics for Human Cancer. Int J Mol Sci 2018; 19:ijms19071979. [PMID: 29986450 PMCID: PMC6073740 DOI: 10.3390/ijms19071979] [Citation(s) in RCA: 605] [Impact Index Per Article: 86.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 06/29/2018] [Accepted: 07/03/2018] [Indexed: 12/17/2022] Open
Abstract
The application of nanotechnology for the treatment of cancer is mostly based on early tumor detection and diagnosis by nanodevices capable of selective targeting and delivery of chemotherapeutic drugs to the specific tumor site. Due to the remarkable properties of gold nanoparticles, they have long been considered as a potential tool for diagnosis of various cancers and for drug delivery applications. These properties include high surface area to volume ratio, surface plasmon resonance, surface chemistry and multi-functionalization, facile synthesis, and stable nature. Moreover, the non-toxic and non-immunogenic nature of gold nanoparticles and the high permeability and retention effect provide additional benefits by enabling easy penetration and accumulation of drugs at the tumor sites. Various innovative approaches with gold nanoparticles are under development. In this review, we provide an overview of recent progress made in the application of gold nanoparticles in the treatment of cancer by tumor detection, drug delivery, imaging, photothermal and photodynamic therapy and their current limitations in terms of bioavailability and the fate of the nanoparticles.
Collapse
|
5
|
Kalimuthu K, Lubin BC, Bazylevich A, Gellerman G, Shpilberg O, Luboshits G, Firer MA. Gold nanoparticles stabilize peptide-drug-conjugates for sustained targeted drug delivery to cancer cells. J Nanobiotechnology 2018; 16:34. [PMID: 29602308 PMCID: PMC5877385 DOI: 10.1186/s12951-018-0362-1] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 03/21/2018] [Indexed: 11/12/2022] Open
Abstract
Background Peptide-drug-conjugates (PDCs) are being developed as an effective strategy to specifically deliver cytotoxic drugs to cancer cells. However one of the challenges to their successful application is the relatively low stability of peptides in the blood, liver and kidneys. Since AuNPs seem to have a longer plasma half-life than PDCs, one approach to overcoming this problem would be to conjugate the PDCs to gold nanoparticles (AuNPs), as these have demonstrated favorable physico-chemical and safety properties for drug delivery systems. We set out to test whether PEG coated-AuNPs could provide a suitable platform for the non-covalent loading of pre-formed PDCs and whether this modification would affect the bioavailability of the PDCs and their cytotoxicity toward target cancer cells. Methods Peptides specifically internalized by A20 murine lymphoma cells were isolated from a phage library displaying 7mer linear peptides. Peptide specificity was validated by flow cytometry and confocal microscopy. PDCs were synthesized containing a selected peptide (P4) and either chlorambucil (Chlor), melphalan (Melph) or bendamustine (Bend). Gold nanoparticles were sequentially coated with citrate, PEG-6000 and then PDC (PDC-PEG-AuNP). The physico-chemical properties of the coated particles were analyzed by electrophoresis, TEM, UV–VIS and FTIR. Stability of free and PDC-coated AuNP was determined. Results Biopanning of the phage library resulted in discovery of several novel peptides that internalized into A20 cells. One of these (P4) was used to synthesize PDCs containing either Chlor, Melph or Bend. All three PDCs specifically killed A20 target cells, however they had short half-lives ranging from 10.6 to 15.4 min. When coated to PEG-AuNPs, the half-lives were extended to 21.0–22.3 h. The PDC-PEG-AuNPs retained cytotoxicity towards the target cells. Moreover, whereas pre-incubation for 24 h of free PDCs almost completely abolished their cytotoxic activity, the PDC-PEG-AuNPs were still active even after 72 h pre-incubation. Conclusions Peptide-drug-conjugates hold potential for improving the target efficacy of chemotherapeutic drugs, however their short half-lives may limit their application. This hurdle can be overcome by easily conjugating them to gold nanoparticles. This conjugation also opens up the possibility of developing slow release formulations of targeted drug delivery systems containing PDCs. Electronic supplementary material The online version of this article (10.1186/s12951-018-0362-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Bat-Chen Lubin
- Dept. Chemical Engineering, Ariel University, 40700, Ariel, Israel.,Eastern R&D Center, Ariel, 40700, Israel
| | | | - Gary Gellerman
- Dept. Chemical Sciences, Ariel University, 40700, Ariel, Israel
| | - Ofer Shpilberg
- Tel Aviv & Pre-Med Faculty, Assuta Medical Center, Institute of Hematology, Ariel University, 40700, Ariel, Israel
| | - Galia Luboshits
- Dept. Chemical Engineering, Ariel University, 40700, Ariel, Israel
| | - Michael A Firer
- Dept. Chemical Engineering, Ariel University, 40700, Ariel, Israel. .,Ariel Center for Applied Cancer Research, Ariel University, 40700, Ariel, Israel.
| |
Collapse
|
6
|
Bashari O, Redko B, Cohen A, Luboshits G, Gellerman G, Firer MA. Discovery of peptide drug carrier candidates for targeted multi-drug delivery into prostate cancer cells. Cancer Lett 2017; 408:164-173. [PMID: 28888997 DOI: 10.1016/j.canlet.2017.08.040] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 11/24/2022]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) remains essentially incurable. Targeted Drug Delivery (TDD) systems may overcome the limitations of current mCRPC therapies. We describe the use of strict criteria to isolate novel prostate cancer cell targeting peptides that specifically deliver drugs into target cells. Phage from a libraries displaying 7mer peptides were exposed to PC-3 cells and only internalized phage were recovered. The ability of these phage to internalize into other prostate cancer cells (LNCaP, DU-145) was validated. The displayed peptides of selected phage clones were synthesized and their specificity for target cells was validated in vitro and in vivo. One peptide (P12) which specifically targeted PC-3 tumors in vivo was incorporated into mono-drug (Chlorambucil, Combretastatin or Camptothecin) and dual-drug (Chlorambucil/Combretastatin or Chlorambucil/Camptothecin) PDCs and the cytotoxic efficacy of these conjugates for target cells was tested. Conjugation of P12 into dual-drug PDCs allowed discovery of new drug combinations with synergistic effects. The use of strict selection criteria can lead to discovery of novel peptides for use as drug carriers for TDD. PDCs represent an effective alternative to current modes of free drug chemotherapy for prostate cancer.
Collapse
Affiliation(s)
- O Bashari
- Dept. Chemical Engineering, Ariel University, Ariel, 40700, Israel.
| | - B Redko
- Dept. Chemical Sciences, Ariel University, Ariel, 40700, Israel.
| | - A Cohen
- Dept. Chemical Engineering, Ariel University, Ariel, 40700, Israel.
| | - G Luboshits
- Dept. Chemical Engineering, Ariel University, Ariel, 40700, Israel.
| | - G Gellerman
- Dept. Chemical Sciences, Ariel University, Ariel, 40700, Israel.
| | - M A Firer
- Dept. Chemical Engineering, Ariel University, Ariel, 40700, Israel.
| |
Collapse
|
7
|
Gilad Y, Firer MA, Rozovsky A, Ragozin E, Redko B, Albeck A, Gellerman G. "Switch off/switch on" regulation of drug cytotoxicity by conjugation to a cell targeting peptide. Eur J Med Chem 2014; 85:139-46. [PMID: 25084142 DOI: 10.1016/j.ejmech.2014.07.073] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/19/2014] [Accepted: 07/21/2014] [Indexed: 11/17/2022]
Abstract
Bi-nuclear amino acid platforms loaded with various drugs for conjugation to a peptide carrier were synthesized using simple and convenient orthogonally protective solid-phase organic synthesis (SPOS). Each arm of the platform carries a different anticancer agent linked through the same or different functional group, providing discrete chemo- and bio-release profiles for each drug, and also enabling "switch off/switch on" regulation of drug cytotoxicity by conjugation to the platform and to a cell targeting peptide. The versatility of this approach enables efficient production of drug-loaded platforms and determination of favorable drug combinations/modes of linkage for subsequent conjugation to a carrier moiety for targeted cancer cell therapy. The results presented here potentiate the application of amino acid platforms for targeted drug delivery (TDD).
Collapse
Affiliation(s)
- Yossi Gilad
- Department of Biological Chemistry, Ariel University, Ariel, 40700, Israel; The Julius Spokojny Bioorganic Chemistry Laboratory, Department of Chemistry, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Michael A Firer
- Department of Chemical Engineering, Ariel University, Ariel, 40700, Israel
| | - Alex Rozovsky
- Department of Biological Chemistry, Ariel University, Ariel, 40700, Israel; The Julius Spokojny Bioorganic Chemistry Laboratory, Department of Chemistry, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Elena Ragozin
- Department of Biological Chemistry, Ariel University, Ariel, 40700, Israel
| | - Boris Redko
- Department of Biological Chemistry, Ariel University, Ariel, 40700, Israel; The Julius Spokojny Bioorganic Chemistry Laboratory, Department of Chemistry, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Amnon Albeck
- The Julius Spokojny Bioorganic Chemistry Laboratory, Department of Chemistry, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Gary Gellerman
- Department of Biological Chemistry, Ariel University, Ariel, 40700, Israel.
| |
Collapse
|
8
|
Drug resistance to chlorambucil in murine B-cell leukemic cells is overcome by its conjugation to a targeting peptide. Anticancer Drugs 2013. [PMID: 23187462 DOI: 10.1097/cad.0b013e32835bb17a] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Targeting drugs through small-molecule carriers with a high affinity to receptors on cancer cells can overcome the lack of target cell specificity of most anticancer drugs. These targeted carrier-drug conjugates are also capable of reversing drug resistance in cancer cells. Although many targeted drug delivery approaches are being tested, the linkage of several and different drugs to a single carrier molecule might further enhance their therapeutic efficacy, particularly if the drugs are engineered for variable time release. This report shows that murine B-cell leukemic cells previously resistant to a chemotherapeutic drug can be made sensitive to that drug as long as it is conjugated to a targeting peptide and, in particular, when the conjugate contains multiple copies of the drug. Using a 13mer peptide (VHFFKNIVTPRTP) derived from the myelin basic protein (p-MBP), dendrimer-based peptide conjugates containing one, two, or four molecules of chlorambucil were synthesized. Although murine hybridomas expressing antibodies to either p-MBP (MBP cells) or a nonrelevant antigen (BCL-1 cells) were both resistant to free chlorambucil, exposure of the cells to the p-MBP-chlorambucil conjugate completely reversed the drug resistance in MBP, but not BCL-1 cells or normal spleen cells. Moreover, at equivalent drug doses, there was significant enhancement in the cytotoxic activity of multidrug versus single-drug copy conjugates. On the basis of these results, the use of multifunctional dendrone linkers bearing several covalently bound cytotoxic agents allows the development of more effective targeted drug systems and enhances the efficacy of currently approved drugs for B-cell leukemia.
Collapse
|
9
|
Grimbaldeston MA, Nakae S, Kalesnikoff J, Tsai M, Galli SJ. Mast cell-derived interleukin 10 limits skin pathology in contact dermatitis and chronic irradiation with ultraviolet B. Nat Immunol 2007; 8:1095-104. [PMID: 17767162 DOI: 10.1038/ni1503] [Citation(s) in RCA: 363] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Accepted: 07/18/2007] [Indexed: 11/08/2022]
Abstract
Allergic contact dermatitis, such as in response to poison ivy or poison oak, and chronic low-dose ultraviolet B irradiation can damage the skin. Mast cells produce proinflammatory mediators that are thought to exacerbate these prevalent acquired immune or innate responses. Here we found that, unexpectedly, mast cells substantially limited the pathology associated with these responses, including infiltrates of leukocytes, epidermal hyperplasia and epidermal necrosis. Production of interleukin 10 by mast cells contributed to the anti-inflammatory or immunosuppressive effects of mast cells in these conditions. Our findings identify a previously unrecognized function for mast cells and mast cell-derived interleukin 10 in limiting leukocyte infiltration, inflammation and tissue damage associated with immunological or innate responses that can injure the skin.
Collapse
Affiliation(s)
- Michele A Grimbaldeston
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305-5176, USA
| | | | | | | | | |
Collapse
|
10
|
Cohen S, Cahan R, Ben-Dov E, Nisnevitch M, Zaritsky A, Firer MA. Specific targeting to murine myeloma cells of Cyt1Aa toxin from Bacillus thuringiensis subspecies israelensis. J Biol Chem 2007; 282:28301-28308. [PMID: 17626007 DOI: 10.1074/jbc.m703567200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Multiple myeloma is currently an incurable cancer of plasma B cells often characterized by overproduction of abnormally high quantities of a patient-specific, clonotypic immunoglobulin "M-protein." The M-protein is expressed on the cell membrane and secreted into the blood. We previously showed that ligand-toxin conjugates (LTC) incorporating the ribosome-inactivating Ricin-A toxin were very effective in specific cytolysis of the anti-ligand antibody-bearing target cells used as models for multiple myeloma. Here, we report on the incorporation of the membrane-disruptive Cyt1Aa toxin from Bacillus thuringiensis subsp. israelensis into LTCs targeted to murine myeloma cells. Proteolytically activated Cyt1Aa was conjugated chemically or genetically through either its amino or carboxyl termini to the major peptidic epitope VHFFKNIVTPRTP (p87-99) of the myelin basic protein. The recombinant fusion-encoding genes were cloned and expressed in acrystalliferous B. thuringiensis subsp. israelensis through the shuttle vector pHT315. Both chemically conjugated and genetically fused LTCs were toxic to anti-myelin basic protein-expressing murine hybridoma cells, but the recombinant conjugates were more active. LTCs comprising the Cyt1Aa toxin might be useful anticancer agents. As a membrane-acting toxin, Cyt1Aa is not likely to induce development of resistant cell lines.
Collapse
Affiliation(s)
- Shmuel Cohen
- Department of Life Sciences, Ben-Gurion University of the Negev, P. O. 653, Be'er-Sheva 84105, Israel; Department of Chemical Engineering and Biotechnology, College of Judea and Samaria, Ariel 44837, Israel
| | - Rivka Cahan
- Department of Chemical Engineering and Biotechnology, College of Judea and Samaria, Ariel 44837, Israel
| | - Eitan Ben-Dov
- Department of Life Sciences, Ben-Gurion University of the Negev, P. O. 653, Be'er-Sheva 84105, Israel; Achva Academic College, MP Shikmim 79800, Israel
| | - Marina Nisnevitch
- Department of Chemical Engineering and Biotechnology, College of Judea and Samaria, Ariel 44837, Israel
| | - Arieh Zaritsky
- Department of Life Sciences, Ben-Gurion University of the Negev, P. O. 653, Be'er-Sheva 84105, Israel
| | - Michael A Firer
- Department of Chemical Engineering and Biotechnology, College of Judea and Samaria, Ariel 44837, Israel.
| |
Collapse
|
11
|
Laptev R, Nisnevitch M, Siboni G, Malik Z, Firer MA. Intracellular chemiluminescence activates targeted photodynamic destruction of leukaemic cells. Br J Cancer 2006; 95:189-96. [PMID: 16819545 PMCID: PMC2360622 DOI: 10.1038/sj.bjc.6603241] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Photodynamic therapy (PDT) involves a two-stage process. A light-absorbing photosensitiser (Ps) is endocytosed and then stimulated by light, inducing transfer of energy to a cytoplasmic acceptor molecule and the generation of reactive oxygen species that initiate damage to cellular membrane components and cytolysis. The expanded use of PDT in the clinic is hindered by the lack of Ps target-cell specificity and the limited tissue penetration by external light radiation. This study demonstrates that bioconjugates composed of transferrin and haematoporphyrin (Tf–Hp), significantly improve the specificity and efficiency of PDT for erythroleukemic cells by a factor of almost seven-fold. Fluorescence microscopy showed that the conjugates accumulate in intracellular vesicles whereas free Hp was mostly membrane bound. Experiments with cells deliberately exposed to Tf–Hp at <LD100 doses showed that surviving cells did not develop resistance to subsequent treatments with the conjugate. Furthermore, we show that the compound luminol induces intracellular chemiluminescence. This strategy was then used to obviate the use of external radiation for Ps activation by incubating the cells with luminol either before or together with Tf–Hp. This novel chemical means of PDT activation induced cytotoxicity in 95% of cells. These combined approaches provide an opportunity to develop broader and more effective applications of PDT.
Collapse
Affiliation(s)
- R Laptev
- Department of Chemical Engineering and Biotechnology, College of Judea and Samaria, Ariel 44837, Israel
| | - M Nisnevitch
- Department of Chemical Engineering and Biotechnology, College of Judea and Samaria, Ariel 44837, Israel
| | - G Siboni
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Z Malik
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - M A Firer
- Department of Chemical Engineering and Biotechnology, College of Judea and Samaria, Ariel 44837, Israel
- E-mail:
| |
Collapse
|