Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 318
1.
Curr Opin Infect Dis ; 37(3): 164-169, 2024 06 01.
Article En | MEDLINE | ID: mdl-38527455

PURPOSE OF REVIEW: Many cholesterol-dependent cytolysin (CDC)-producing pathogens pose a significant threat to human health. Herein, we review the pore-dependent and -independent properties CDCs possess to assist pathogens in evading the host immune response. RECENT FINDINGS: Within the last 5 years, exciting new research suggests CDCs can act to inhibit important immune functions, disrupt critical cell signaling pathways, and have tissue-specific effects. Additionally, recent studies have identified a key region of CDCs that generates robust immunity, providing resources for the development of CDC-based vaccines. SUMMARY: This review provides new information on how CDCs alter host immune responses to aid bacteria in pathogenesis. These studies can assist in the design of more efficient vaccines and therapeutics against CDCs that will enhance the immune response to CDC-producing pathogens while mitigating the dampening effects CDCs have on the host immune response.


Cholesterol , Cytotoxins , Humans , Cholesterol/metabolism , Cytotoxins/immunology , Host-Pathogen Interactions/immunology , Bacteria/immunology , Immune Evasion/immunology
2.
PLoS Negl Trop Dis ; 15(10): e0009841, 2021 10.
Article En | MEDLINE | ID: mdl-34634067

Development of a rapid, on-site detection tool for snakebite is highly sought after, owing to its clinically and forensically relevant medicolegal significance. Polyvalent antivenom therapy in the management of such envenomation cases is finite due to its poor venom neutralization capabilities as well as diagnostic ramifications manifested as untoward immunological reactions. For precise molecular diagnosis of elapid venoms of the big four snakes, we have developed a lateral flow kit using a monoclonal antibody (AB1; IgG1 - κ chain; Kd: 31 nM) generated against recombinant cytotoxin-7 (rCTX-7; 7.7 kDa) protein of the elapid venom. The monoclonal antibody specifically detected the venoms of Naja naja (p < 0.0001) and Bungarus caeruleus (p<0.0001), without showing any immunoreactivity against the viperidae snakes in big four venomous snakes. The kit developed attained the limit of quantitation of 170 pg/µL and 2.1 ng/µL in spiked buffer samples and 28.7 ng/µL and 110 ng/µL in spiked serum samples for detection of N. naja and B. caeruleus venoms, respectively. This kit holds enormous potential in identification of elapid venom of the big four snakes for effective prognosis of an envenomation; as per the existing medical guidelines.


Colorimetry/methods , Cytotoxins/analysis , Elapidae/immunology , Immunoassay/methods , Immunotoxins/analysis , Snake Venoms/analysis , Animals , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/immunology , Bungarus/genetics , Bungarus/physiology , Cytotoxins/genetics , Cytotoxins/immunology , Elapid Venoms/analysis , Elapid Venoms/genetics , Elapid Venoms/immunology , Elapidae/physiology , Immunotoxins/genetics , Immunotoxins/immunology , Naja naja/immunology , Naja naja/physiology , Snake Venoms/immunology , Viperidae/immunology , Viperidae/physiology
3.
Methods Mol Biol ; 2033: 1-14, 2019.
Article En | MEDLINE | ID: mdl-31332743

Antibody-drug conjugates (ADCs) have been proven to be a successful therapeutic concept, allowing targeted delivery of highly potent active pharmaceutical ingredients (HPAPIs) selectively to tumor tissue. So far, HPAPIs have been mainly attached to the antibody via a chemical reaction of the payload with lysine or cysteine side chains of the antibody backbone. However, these conventional conjugation technologies result in formation of rather heterogeneous products with undesired properties. To overcome the limitations of heterogeneous ADC mixtures, several site-specific conjugation technologies have been developed over the last years. Originally pioneered by scientist from Genentech with their work on THIOMABs, several engineered cysteine mAb ADCs (ECM-ADCs) are now investigated in clinical trials. Here, we describe in detail how to engineer additional cysteines into antibodies and efficiently use them as highly site-specific conjugation sites for HPAPIs.


Antibodies, Monoclonal, Humanized/genetics , Cytotoxins/genetics , Immunoconjugates/genetics , Protein Engineering , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological , Cell Proliferation/drug effects , Cysteine/chemistry , Cysteine/genetics , Cytotoxins/chemistry , Cytotoxins/immunology , Cytotoxins/pharmacology , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunoconjugates/pharmacology , Mice , Mutagenesis, Site-Directed , Sulfhydryl Compounds/chemistry , Trastuzumab/chemistry , Trastuzumab/genetics , Trastuzumab/immunology , Xenograft Model Antitumor Assays
4.
Curr Issues Mol Biol ; 32: 645-700, 2019.
Article En | MEDLINE | ID: mdl-31166182

Streptococcus pneumoniae (Spn) and Streptococcus pyogenes (Spy) cause many invasive and noninvasive diseases responsible for high morbidity and mortality worldwide. Safe, efficacious and affordable vaccines could have a significant, positive impact on the global infectious disease burden. Since the implementation of pneumococcal vaccine in the 1980s, the incidence of Spn infection has decreased significantly. Still so, these currently used multivalent polysaccharides and conjugated pneumococcal vaccines have some limitations. For Spy, there are even no vaccines available yet. There is an urgent need of new vaccines against Spn and Spy. Encouragingly, with the hard work of many investigators worldwide, a number of new vaccines candidates are developed with promising results. Of them, many have already entered the clinical trial stage. This review will describe the current status of Spn and Spy vaccine development, with particular focus on protein-based strategy.


Bacterial Proteins/immunology , Immunogenicity, Vaccine , Polysaccharides, Bacterial/immunology , Streptococcal Infections/prevention & control , Streptococcal Vaccines/biosynthesis , Streptococcus pneumoniae/drug effects , Bacterial Proteins/genetics , Clinical Trials as Topic , Cytotoxins/genetics , Cytotoxins/immunology , Fimbriae, Bacterial/chemistry , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/immunology , Gene Expression , Humans , Polysaccharides, Bacterial/chemistry , Serogroup , Streptococcal Infections/immunology , Streptococcal Infections/pathology , Streptococcal Vaccines/administration & dosage , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/pathogenicity , Vaccines, Attenuated , Vaccines, Conjugate , Vaccines, Subunit , Virulence
5.
Article En | MEDLINE | ID: mdl-31138568

ASN100 is a novel antibody combination of two fully human IgG1(κ) monoclonal antibodies (MAbs), ASN-1 and ASN-2, which neutralize six Staphylococcus aureus cytotoxins, alpha-hemolysin (Hla) and five bicomponent leukocidins. We assessed the safety, tolerability, and serum and lung pharmacokinetics of ASN100 in a randomized, double-blind, placebo-controlled single-dose-escalation first-in-human study. Fifty-two healthy volunteers were enrolled and randomized to receive either ASN-1, ASN-2, a combination of both MAbs (ASN100), or a corresponding placebo. Thirty-two subjects in the double-blind dose escalation portion of the study received ASN-1 or ASN-2 at a 200-, 600-, 1,800-, or 4,000-mg dose, or placebo. Eight subjects received both MAbs simultaneously in a 1:1 ratio (ASN100) at 3,600 or 8,000 mg, or they received placebos. Twelve additional subjects received open-label ASN100 at 3,600 or 8,000 mg to assess the pharmacokinetics of ASN-1 and ASN-2 in epithelial lining fluid (ELF) by bronchoalveolar lavage fluid sampling. Subjects were monitored for 98 days (double-blind cohorts) or 30 days (open-label cohorts) for safety assessment. No dose-limiting toxicities were observed, and all adverse events were mild and transient, with only two adverse events considered possibly related to the investigational product. ASN100 exhibited linear serum pharmacokinetics with a half-life of approximately 3 weeks and showed detectable penetration into the ELF. No treatment-emergent anti-drug antibody responses were detected. The toxin neutralizing potency of ASN100 in human serum was confirmed up to 58 days postdosing. The favorable safety profile, ELF penetration, and maintained functional activity in serum supported the further clinical development of ASN100.


Anti-Bacterial Agents/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Bacterial Toxins/antagonists & inhibitors , Cytotoxins/immunology , Adult , Anti-Bacterial Agents/pharmacokinetics , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Bronchoalveolar Lavage Fluid , Cytotoxins/antagonists & inhibitors , Cytotoxins/metabolism , Double-Blind Method , Female , Healthy Volunteers , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/immunology , Humans , Leukocidins/antagonists & inhibitors , Leukocidins/immunology , Male , Placebos , Staphylococcal Infections , Staphylococcus aureus/immunology
6.
Sci Rep ; 8(1): 6458, 2018 04 24.
Article En | MEDLINE | ID: mdl-29691463

Necrotizing soft tissue infections are lethal polymicrobial infections. Two key microbes that cause necrotizing soft tissue infections are Streptococcus pyogenes and Clostridium perfringens. These pathogens evade innate immunity using multiple virulence factors, including cholesterol-dependent cytolysins (CDCs). CDCs are resisted by mammalian cells through the sequestration and shedding of pores during intrinsic membrane repair. One hypothesis is that vesicle shedding promotes immune evasion by concomitantly eliminating key signaling proteins present in cholesterol-rich microdomains. To test this hypothesis, murine macrophages were challenged with sublytic CDC doses. CDCs suppressed LPS or IFNγ-stimulated TNFα production and CD69 and CD86 surface expression. This suppression was cell intrinsic. Two membrane repair pathways, patch repair and intrinsic repair, might mediate TNFα suppression. However, patch repair did not correlate with TNFα suppression. Intrinsic repair partially contributed to macrophage dysfunction because TLR4 and the IFNγR were partially shed following CDC challenge. Intrinsic repair was not sufficient for suppression, because pore formation was also required. These findings suggest that even when CDCs fail to kill cells, they may impair innate immune signaling responses dependent on cholesterol-rich microdomains. This is one potential mechanism to explain the lethality of S. pyogenes and C. perfringens during necrotizing soft tissue infections.


Cytotoxins/metabolism , Cytotoxins/pharmacology , Macrophages/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , B7-2 Antigen/metabolism , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Cell Membrane/metabolism , Cholesterol/metabolism , Clostridium perfringens/metabolism , Clostridium perfringens/pathogenicity , Cytotoxins/immunology , Female , Hemolysin Proteins/metabolism , Immunity, Innate/immunology , Immunity, Innate/physiology , Lectins, C-Type/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/drug effects , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/pathogenicity , Tumor Necrosis Factor-alpha/drug effects
7.
J Biomed Sci ; 24(1): 58, 2017 Aug 19.
Article En | MEDLINE | ID: mdl-28822352

BACKGROUND: Vibrio vulnificus is a marine bacterial species that causes opportunistic infections manifested by serious skin lesions and fulminant septicemia in humans. We have previously shown that the multifunctional autoprocessing repeats in toxin (MARTXVv1) of a biotype 1 V. vulnificus strain promotes survival of this organism in the host by preventing it from engulfment by the phagocytes. The purpose of this study was to further explore how MARTXVv1 inhibits phagocytosis of this microorganism by the macrophage. METHODS: We compared between a wild-type V. vulnificus strain and its MARTXVv1-deficient mutant for a variety of phagocytosis-related responses, including morphological change and activation of signaling molecules, they induced in the macrophage. We also characterized a set of MARTXVv1 domain-deletion mutants to define the regions associated with antiphagocytosis activity. RESULTS: The RAW 264.7 cells and mouse peritoneal exudate macrophages underwent cell rounding accompanied by F-actin disorganization in the presence of MARTXVv1. In addition, phosphorylation of some F-actin rearrangement-associated signaling molecules, including Lyn, Fgr and Hck of the Src family kinases (SFKs), focal adhesion kinase (FAK), proline-rich tyrosine kinase 2 (Pyk2), phosphoinositide 3-kinase (PI3K) and Akt, but not p38, was decreased. By using specific inhibitors, we found that these kinases were all involved in the phagocytosis of MARTXVv1-deficient mutant in an order of SFKs-FAK/Pyk2-PI3K-Akt. Deletion of the effector domains in the central region of MARTXVv1 could lead to reduced cytotoxicity, depending on the region and size of deletion, but did not affect the antiphagocytosis activity and ability to cause rounding of macrophage. Reduced phosphorylation of Akt was closely associated with inhibition of phagocytosis by the wild-type strain and MARTXVv1 domain-deletion mutants, and expression of the constitutively active Akt, myr-Akt, enhanced the engulfment of these strains by macrophage. CONCLUSIONS: MARTXVv1 could inactivate the SFKs-FAK/Pyk2-PI3K-Akt signaling pathway in the macrophages. This might lead to impaired phagocytosis of the V. vulnificus-infected macrophage. The majority of the central region of MARTXVv1 is not associated with the antiphagocytosis activity.


Bacterial Toxins/immunology , Phagocytosis/immunology , Vibrio Infections/microbiology , Vibrio vulnificus/immunology , Vibrio vulnificus/pathogenicity , Animals , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Cytotoxins/immunology , Cytotoxins/metabolism , Macrophages/immunology , Male , Mice, Inbred BALB C , Vibrio Infections/pathology , Vibrio vulnificus/genetics
8.
Immunity ; 45(6): 1258-1269, 2016 12 20.
Article En | MEDLINE | ID: mdl-27939674

Programmed death and shedding of epithelial cells is a powerful defense mechanism to reduce bacterial burden during infection but this activity cannot be indiscriminate because of the critical barrier function of the epithelium. We report that during cystitis, shedding of infected bladder epithelial cells (BECs) was preceded by the recruitment of mast cells (MCs) directly underneath the superficial epithelium where they docked and extruded their granules. MCs were responding to interleukin-1ß (IL-1ß) secreted by BECs after inflammasome and caspase-1 signaling. Upon uptake of granule-associated chymase (mouse MC protease 4 [mMCPT4]), BECs underwent caspase-1-associated cytolysis and exfoliation. Thus, infected epithelial cells require a specific cue for cytolysis from recruited sentinel inflammatory cells before shedding.


Chymases/immunology , Cytotoxins/immunology , Epithelial Cells/microbiology , Mast Cells/immunology , Urinary Tract Infections/immunology , Animals , Cell Degranulation/immunology , Cell Line , Cytoplasmic Granules/chemistry , Female , Fluorescent Antibody Technique , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
Clin. transl. oncol. (Print) ; 18(7): 653-659, jul. 2016.
Article En | IBECS | ID: ibc-153488

Historically, patients diagnosed with metastatic pancreatic cancer have faced a grim prognosis. The survival benefit seen with systemic chemotherapies and even combinations thereof have been disappointing. However, growing data suggest that the microenvironment of pancreatic cancer may be contributing to this poor prognosis. This microenvironment has a dense fibrotic stroma, and is hypoxic and highly immunosuppressive, all of which pose barriers to treatment. Newer strategies looking to disrupt the fibrotic stroma, target hypoxic areas, and improve local immune responses in the tumor microenvironment are currently undergoing clinical evaluation and seem to offer great promise. In addition to these therapies, preclinical work evaluating novel cytotoxic agents including nanoparticles has also been encouraging. While much research still needs to be done, these strategies offer new hope for patients with pancreatic cancer (AU)


No disponible


Humans , Male , Female , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , Prognosis , Immunosuppression Therapy/instrumentation , Immunosuppression Therapy/methods , Nanoparticles/therapeutic use , Cytotoxins/immunology , Cytotoxins/therapeutic use
10.
J Cyst Fibros ; 15(5): 597-604, 2016 09.
Article En | MEDLINE | ID: mdl-26821814

BACKGROUND: Staphylococcus aureus is one of the earliest bacterial pathogens to colonize the lungs of children with cystic fibrosis and is an important contributor to pulmonary exacerbations. The adaptive host response to S. aureus in cystic fibrosis remains inadequately defined and has important implications for pathogenesis and potential interventions. The objectives of this study were to determine the functional antibody response to select staphylococcal exotoxins (LukAB, alpha-hemolysin, and PVL) in children with cystic fibrosis and to evaluate the relationship of this response with pulmonary exacerbations. METHODS: Fifty children with cystic fibrosis were enrolled and followed prospectively for 12months. Clinical characteristics and serologic profiles were assessed at routine visits and during pulmonary exacerbations, and functional antibody assessments were performed to measure neutralization of LukAB-mediated cytotoxicity. RESULTS: For each antigen, geometric mean titers were significantly higher if S. aureus was detected at the time of exacerbation. For LukAB, geometric mean titers were significantly higher at exacerbation follow-up compared to titers during the exacerbation, consistent with expression during human disease, and the humoral response capably neutralized LukAB-mediated cytotoxicity. Moreover, the presence of a positive S. aureus culture during a pulmonary exacerbation was associated with 31-fold higher odds of having a LukA titer ≥1:160, suggesting potential diagnostic capability of this assay. CONCLUSIONS: The leukotoxin LukAB is expressed by S. aureus and recognized by the human adaptive immune response in the setting of pulmonary infection in cystic fibrosis. Anti-LukAB antibodies were not only predictive of positive staphylococcal culture during exacerbation, but also functional in the neutralization of this toxin.


Bacterial Proteins/immunology , Cystic Fibrosis , Leukocidins/immunology , Staphylococcal Infections , Staphylococcus aureus , Adaptive Immunity/immunology , Adolescent , Antibody Formation/immunology , Child , Child, Preschool , Cystic Fibrosis/immunology , Cystic Fibrosis/microbiology , Cystic Fibrosis/physiopathology , Cystic Fibrosis/therapy , Cytotoxins/immunology , Female , Humans , Lung Diseases/diagnosis , Lung Diseases/immunology , Lung Diseases/physiopathology , Male , Prospective Studies , Staphylococcal Infections/diagnosis , Staphylococcal Infections/immunology , Staphylococcal Infections/physiopathology , Staphylococcus aureus/immunology , Staphylococcus aureus/isolation & purification , Symptom Flare Up , United States
11.
J Immunol ; 195(7): 3190-7, 2015 Oct 01.
Article En | MEDLINE | ID: mdl-26297758

The discovery of new vaccines against infectious diseases and cancer requires the development of novel adjuvants with well-defined activities. The TLR4 agonist adjuvant GLA-SE elicits robust Th1 responses to a variety of vaccine Ags and is in clinical development for both infectious diseases and cancer. We demonstrate that immunization with a recombinant protein Ag and GLA-SE also induces granzyme A expression in CD4 T cells and produces cytolytic cells that can be detected in vivo. Surprisingly, these in vivo CTLs were CD4 T cells, not CD8 T cells, and this cytolytic activity was not dependent on granzyme A/B or perforin. Unlike previously reported CD4 CTLs, the transcription factors Tbet and Eomes were not necessary for their development. CTL activity was also independent of the Fas ligand-Fas, TRAIL-DR5, and canonical death pathways, indicating a novel mechanism of CTL activity. Rather, the in vivo CD4 CTL activity induced by vaccination required T cell expression of CD154 (CD40L) and target cell expression of CD40. Thus, vaccination with a TLR4 agonist adjuvant induces CD4 CTLs, which kill through a previously unknown CD154-dependent mechanism.


CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , CD40 Ligand/immunology , T-Lymphocytes, Cytotoxic/immunology , Adjuvants, Immunologic/pharmacology , Animals , CD40 Antigens/biosynthesis , CD40 Ligand/biosynthesis , Cytotoxins/immunology , Fas Ligand Protein/immunology , Granzymes/biosynthesis , Granzymes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , T-Box Domain Proteins/immunology , TNF-Related Apoptosis-Inducing Ligand/immunology , Th1 Cells/immunology , Vaccination
12.
Nanomedicine (Lond) ; 10(13): 2075-92, 2015.
Article En | MEDLINE | ID: mdl-26135328

Zinc oxide nanoparticles (ZnO NPs) have useful physicochemical advantages, and are used extensively. This has raised concerns regarding their potential toxicity. ZnO NP attributes that contribute to cytotoxicity and immune reactivity, however, seem to vary across literature considerably. Largely, dissolution and generation of reactive oxygen species appear to be the most commonly reported paradigms. Moreover, ZnO NP size and shape may also contribute toward their overall nano-bio interactions. Analysis is further complicated by factors such as adsorption of proteins on the NP surface, which may influence their bioreactivity. The main aim of this review is to give a systematic overview of the postulates explaining cytotoxic, inflammatory and genotoxic effects of ZnO NPs when exposed to different types of cells in vitro and in vivo.


Cytotoxins/toxicity , Inflammation/chemically induced , Mutagens/toxicity , Nanoparticles/toxicity , Zinc Oxide/toxicity , Animals , Apoptosis/drug effects , Cytotoxins/chemistry , Cytotoxins/immunology , Cytotoxins/metabolism , Humans , Immunologic Factors/chemistry , Immunologic Factors/immunology , Immunologic Factors/metabolism , Immunologic Factors/toxicity , Inflammation/immunology , Inflammation/metabolism , Mutagens/chemistry , Mutagens/metabolism , Nanoparticles/chemistry , Nanoparticles/metabolism , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Zinc Oxide/chemistry , Zinc Oxide/immunology , Zinc Oxide/metabolism
13.
Nat Rev Drug Discov ; 14(7): 487-98, 2015 Jul.
Article En | MEDLINE | ID: mdl-26000725

Scientific insights into the human immune system have recently led to unprecedented breakthroughs in immunotherapy. In the twenty-first century, drugs and cell-based therapies developed to bolster humoral and T cell immunity represent an established and growing component of cancer therapeutics. Although natural killer (NK) cells have long been known to have advantages over T cells in terms of their capacity to induce antigen-independent host immune responses against malignancies, their therapeutic potential in the clinic has been largely unexplored. A growing number of scientific discoveries into pathways that both activate and suppress NK cell function, as well as methods to sensitize tumours to NK cell cytotoxicity, have led to the development of numerous pharmacological and genetic methods to enhance NK cell antitumour immunity. These findings, as well as advances in our ability to expand NK cells ex vivo and manipulate their capacity to home to the tumour, have now provided investigators with a variety of new methods and strategies to harness the full potential of NK cell-based cancer immunotherapy in the clinic.


Immunotherapy/methods , Killer Cells, Natural/immunology , Neoplasms/immunology , Neoplasms/therapy , Animals , Biological Products/immunology , Biological Products/therapeutic use , Cytotoxins/immunology , Cytotoxins/therapeutic use , Humans , Immunotherapy/trends
14.
Curr Top Med Chem ; 14(24): 2822-34, 2015.
Article En | MEDLINE | ID: mdl-25487009

Antibody-drug conjugates (ADCs) are an emerging area of study within medicinal chemistry and are thought of as sophisticated drug delivery systems due to their specificity to a disease-targeted antigen. ADCs have been actively utilized as therapeutics for hematological and solid tumor cancers due to their capability to deliver a cytotoxic compound to a specific cancer cell without affecting normal cells. An antibody drug conjugate has three major constituents: a monoclonal antibody (mAb), a chemical linker, and a potent cytotoxic payload. There has been a continuing effort to optimize antibody-drug conjugates, with the primary focus of design and development directed at either the mAb or the chemical linker, with little effort devoted to the optimization of payload compounds. In fact, among the 114 ongoing or recently completed clinical trials, there is generally a lack of diversity in the cytotoxic payloads that are utilized, with only seven payload compounds reported (four additional trials are ongoing with structures that have not been reported). Six of these seven payload compounds are derived from natural product sources, highlighting the importance of natural products as cytotoxic payloads for ADC.


Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemistry , Biological Products/chemistry , Cytotoxins/chemistry , Hematologic Neoplasms/drug therapy , Immunoconjugates/chemistry , Aminoglycosides/chemistry , Aminoglycosides/immunology , Aminoglycosides/therapeutic use , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, Neoplasm/immunology , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Brentuximab Vedotin , Clinical Trials as Topic , Cytotoxins/immunology , Cytotoxins/therapeutic use , Drug Delivery Systems , Gemtuzumab , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Humans , Immunoconjugates/immunology , Immunoconjugates/therapeutic use , Molecular Targeted Therapy , Protein Binding , Trastuzumab
15.
J Pharmacokinet Pharmacodyn ; 41(5): 461-78, 2014 Oct.
Article En | MEDLINE | ID: mdl-25281420

Cancer therapies that harness the actions of the immune response, such as targeted monoclonal antibody treatments and therapeutic vaccines, are relatively new and promising in the landscape of cancer treatment options. Mathematical modeling and simulation of immune-modifying therapies can help to offset the costs of drug discovery and development, and encourage progress toward new immunotherapies. Despite advances in cancer immunology research, questions such as how the immune system interacts with a growing tumor, and which components of the immune system play significant roles in responding to immunotherapy are still not well understood. Mathematical modeling and simulation are powerful tools that provide an analytical framework in which to address such questions. A quantitative understanding of the kinetics of the immune response to treatment is crucial in designing treatment strategies, such as dosing, timing, and predicting the response to a specific treatment. These models can be used both descriptively and predictively. In this chapter, various mathematical models that address different cancer treatments, including cytotoxic chemotherapy, immunotherapy, and combinations of both treatments, are presented. The aim of this chapter is to highlight the importance of mathematical modeling and simulation in the design of immunotherapy protocols for cancer treatment. The results demonstrate the power of these approaches in explaining determinants that are fundamental to cancer-immune dynamics, therapeutic success, and the development of efficient therapies.


Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Cytotoxins/immunology , Cytotoxins/therapeutic use , Immunotherapy , Models, Immunological , Neoplasms/drug therapy , Neoplasms/immunology , Computer Simulation , Humans
16.
Toxins (Basel) ; 6(3): 950-72, 2014 Mar 04.
Article En | MEDLINE | ID: mdl-24599233

Staphylococccus aureus represents one of the most challenging human pathogens as well as a common colonizer of human skin and mucosal surfaces. S. aureus causes a wide range of diseases from skin and soft tissue infection (SSTI) to debilitating and life-threatening conditions such as osteomyelitis, endocarditis, and necrotizing pneumonia. The range of diseases reflects the remarkable diversity of the virulence factors produced by this pathogen, including surface antigens involved in the establishment of infection and a large number of toxins that mediate a vast array of cellular responses. The staphylococcal toxins are generally believed to have evolved to disarm the innate immune system, the first line of defense against this pathogen. This review focuses on recent advances on elucidating the biological functions of S. aureus bicomponent pore-forming toxins (BCPFTs) and their utility as targets for preventive and therapeutic intervention. These toxins are cytolytic to a variety of immune cells, primarily neutrophils, as well as cells with a critical barrier function. The lytic activity of BCPFTs towards immune cells implies a critical role in immune evasion, and a number of epidemiological studies and animal experiments relate these toxins to clinical disease, particularly SSTI and necrotizing pneumonia. Antibody-mediated neutralization of this lytic activity may provide a strategy for development of toxoid-based vaccines or immunotherapeutics for prevention or mitigation of clinical diseases. However, certain BCPFTs have been proposed to act as danger signals that may alert the immune system through an inflammatory response. The utility of a neutralizing vaccination strategy must be weighed against such immune-activating potential.


Bacterial Toxins/immunology , Cytotoxins/immunology , Pore Forming Cytotoxic Proteins/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus , Animals , Bacterial Toxins/metabolism , Cytotoxins/metabolism , Humans , Immunotherapy , Pore Forming Cytotoxic Proteins/metabolism , Protein Subunits/immunology , Protein Subunits/metabolism , Staphylococcal Infections/prevention & control , Staphylococcal Infections/therapy , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity
17.
Infect Immun ; 82(3): 1234-42, 2014 Mar.
Article En | MEDLINE | ID: mdl-24379282

Despite the importance of Staphylococcus aureus as a common invasive bacterial pathogen, the humoral response to infection remains inadequately defined, particularly in children. The purpose of this study was to assess the humoral response to extracellular staphylococcal virulence factors, including the bicomponent leukotoxins, which are critical for the cytotoxicity of S. aureus toward human neutrophils. Children with culture-proven S. aureus infection were prospectively enrolled and stratified by disease type. Fifty-three children were enrolled in the study, of which 90% had invasive disease. Serum samples were obtained during the acute (within 48 h) and convalescent (4 to 6 weeks postinfection) phases, at which point both IgG titers against S. aureus exotoxins were determined, and the functionality of the generated antibodies was evaluated. Molecular characterization of clinical isolates was also performed. We observed a marked rise in antibody titer from acute-phase to convalescent-phase sera for LukAB, the most recently described S. aureus bicomponent leukotoxin. LukAB production by the isolates was strongly correlated with cytotoxicity in vitro, and sera containing anti-LukAB antibodies potently neutralized cytotoxicity. Antibodies to S. aureus antigens were detectable in healthy pediatric controls but at much lower titers than in sera from infected subjects. The discovery of a high-titer, neutralizing antibody response to LukAB during invasive infections suggests that this toxin is produced in vivo and that it elicits a functional humoral response.


Antibodies, Neutralizing/immunology , Bacterial Proteins/immunology , Cytotoxins/immunology , Leukocidins/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Child , Female , Humans , Male , Virulence Factors/immunology
18.
Mol Oncol ; 8(2): 378-88, 2014 Mar.
Article En | MEDLINE | ID: mdl-24389243

In clinical practice, targeted therapies are usually administered together with chemotherapeutics. However, little is known whether conventional cytotoxic agents enhance the efficacy of targeted compounds, and whether a possible synergy would be dictated by drug-sensitizing genetic alterations. To explore these issues, we leveraged the design of clinical studies in humans to conduct a multi-arm trial in an 'in-cell' format. Using the MET oncogene as a model target and a panel of genetically characterized cell lines as a reference population, we found that two different chemotherapeutic regimens - cisplatin and 5-fluorouracil - exerted widespread cytotoxic activity that was not further enhanced by MET inhibition with a monovalent anti-MET antibody. From a complementary perspective, targeted MET inhibition was successful in a selected complement of cells harboring MET genomic lesions. In this latter setting, addition of chemotherapy did not provide a therapeutic advantage. Mechanistically, chemotherapeutics did not influence the basal activity of MET in cells with normal MET genomic status nor did they contribute to neutralize MET signals in cells with MET amplification. These data suggest that tumors displaying MET aberrations achieve plateau responses by MET monotherapy and do not receive further benefit by addition of cytotoxic treatments.


Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/pharmacology , Cytotoxins/pharmacology , Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , Cytotoxins/immunology , Drug Screening Assays, Antitumor , Fluorouracil/pharmacology , Humans , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Proto-Oncogene Proteins c-met/genetics
19.
Clin Vaccine Immunol ; 21(2): 181-7, 2014 Feb.
Article En | MEDLINE | ID: mdl-24334685

Infectious bovine keratoconjunctivitis (IBK) caused by Moraxella bovis is the most common eye disease of cattle. The pathogenesis of M. bovis requires the expression of pili that enable the organism to attach to the ocular surface and an RTX (repeats in the structural toxin) toxin (cytotoxin or hemolysin), which is cytotoxic to corneal epithelial cells. In this pilot study, ocular mucosal immune responses of steers were measured following intranasal (i.n.) vaccination with a recombinant M. bovis cytotoxin adjuvanted with polyacrylic acid. Beef steers were vaccinated with either 500 µg (n = 3) or 200 µg (n = 3) of recombinant M. bovis cytotoxin plus adjuvant. Control group steers (n = 2) were vaccinated with adjuvant alone, and all steers were given a booster on day 21. Antigen-specific tear IgA and tear IgG, tear cytotoxin-neutralizing antibody responses, and serum cytotoxin-neutralizing antibody responses were determined in samples collected prevaccination and on days 14, 28, 42, and 55. Changes in tear antigen-specific IgA levels from day 0 to days 28, 42, and 55 were significantly different between groups; however, in post hoc comparisons between individual group pairs at the tested time points, the differences were not significant. Our results suggest that i.n. vaccination of cattle with recombinant M. bovis cytotoxin adjuvanted with polyacrylic acid effects changes in ocular antigen-specific IgA concentrations. The use of intranasally administered recombinant M. bovis cytotoxin adjuvanted with polyacrylic acid could provide an alternative to parenteral vaccination of cattle for immunoprophylaxis against IBK.


Bacterial Vaccines/immunology , Cattle Diseases/prevention & control , Cytotoxins/immunology , Eye/immunology , Keratoconjunctivitis, Infectious/prevention & control , Moraxella bovis/immunology , Acrylic Resins/administration & dosage , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibodies, Bacterial/analysis , Antibodies, Bacterial/blood , Antibodies, Neutralizing/analysis , Antibodies, Neutralizing/blood , Bacterial Vaccines/administration & dosage , Cattle , Cytotoxins/administration & dosage , Immunoglobulin A/analysis , Immunoglobulin G/analysis , Immunoglobulin G/blood , Tears/immunology , Vaccination/methods
20.
J Infect Dis ; 209(12): 1955-62, 2014 Jun 15.
Article En | MEDLINE | ID: mdl-24357631

BACKGROUND: Staphylococcus aureus causes serious infections in both hospital and community settings. Attempts have been made to prevent human infection through vaccination against bacterial cell-surface antigens; thus far all have failed. Here we show that superantigens and cytolysins, when used in vaccine cocktails, provide protection from S. aureus USA100-USA400 intrapulmonary challenge. METHODS: Rabbits were actively vaccinated (wild-type toxins or toxoids) or passively immunized (hyperimmune serum) against combinations of superantigens (toxic shock syndrome toxin 1, enterotoxins B and C, and enterotoxin-like X) and cytolysins (α-, ß-, and γ-toxins) and challenged intrapulmonarily with multiple strains of S. aureus, both methicillin-sensitive and methicillin-resistant. RESULTS: Active vaccination against a cocktail containing bacterial cell-surface antigens enhanced disease severity as tested by infective endocarditis. Active vaccination against secreted superantigens and cytolysins resulted in protection of 86 of 88 rabbits when challenged intrapulmonarily with 9 different S. aureus strains, compared to only 1 of 88 nonvaccinated animals. Passive immunization studies demonstrated that production of neutralizing antibodies was an important mechanism of protection. CONCLUSIONS: The data suggest that vaccination against bacterial cell-surface antigens increases disease severity, but vaccination against secreted virulence factors provides protection against S. aureus. These results advance our understanding of S. aureus pathogenesis and have important implications in disease prevention.


Immunization, Passive , Pneumonia, Staphylococcal/prevention & control , Staphylococcal Vaccines/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Bacterial Toxins/immunology , Cytotoxins/immunology , Disease Models, Animal , Endocarditis, Bacterial/immunology , Endocarditis, Bacterial/prevention & control , Enterotoxins/immunology , Female , Male , Methicillin-Resistant Staphylococcus aureus/immunology , Pneumonia, Staphylococcal/immunology , Rabbits , Superantigens/immunology , Virulence Factors/immunology
...