Subject(s)
Antivenins/therapeutic use , Snake Bites/therapy , Snake Venoms/adverse effects , Animals , Humans , Nervous System Diseases/chemically induced , Neurotoxins/adverse effects , Snake Bites/complications , Snake Bites/epidemiology , Snake Bites/physiopathology , Snake Venoms/antagonists & inhibitors , Snakes/anatomy & histologyABSTRACT
OBJECTIVE: Snake envenomation has been poorly studied in developing countries. 'Early morning neuroparalytic syndrome' (EMNS), the classical clinical constellation caused by krait bites, refers to nighttime, indoor bites where nonspecific symptoms progress to neuroparalysis. Literature regarding EMNS in children is scarce. This study was planned to describe the clinical profile, intensive care needs and predictors of outcome in children with EMNS. METHODS: It is a retrospective study of children below 12 years admitted with a clinical diagnosis of snake envenomation to the pediatric intensive care unit (PICU) of a tertiary care hospital in North India. Patient records were reviewed from the electronic patient database manager. Comparison was made between the EMNS group and the non-EMNS group and between survivors and nonsurvivors within the EMNS group. RESULTS: Of the 111 children with snake envenomation, 76 had neuroparalysis (68%) and 51 had EMNS. In the EMNS cohort, 37 (72.5%) belonged to rural areas, 46 (90.2%) had indoor bites and 39 (76.5%) were witnessed. Patients with EMNS were more likely to have absent fang marks, hypoxemia at admission, bulbar palsy and need for PICU admission. Mortality rate was 13.7% in EMNS; predictors included younger age, presence of ptosis, cardiac arrest at admission and nonavailability of PICU bed (univariable analysis) but none of them independently predicted mortality. CONCLUSION: Younger age, presence of ptosis, cardiac arrest at admission and nonavailability of intensive care beds increase the risk of mortality in children with EMNS. Timely recognition and respiratory support may reduce mortality in these children.
Subject(s)
Neurotoxicity Syndromes/diagnosis , Paralysis/etiology , Snake Bites/complications , Snake Venoms/adverse effects , Child , Child, Preschool , Female , Humans , Intensive Care Units, Pediatric , Male , Mortality , Neurotoxicity Syndromes/epidemiology , Paralysis/therapy , Snake Bites/diagnosis , Snake Bites/mortality , Snake Bites/therapyABSTRACT
BACKGROUND: Coral snake bites from Micrurus fulvius and Micrurus tener account for < 1% of all snake bites in North America. Coral snake envenomation may cause significant neurotoxicity, including respiratory insufficiency, and its onset may be delayed up to 13 h. CASE REPORT: We present a unique patient encounter of M. tener venom exposure through the ocular mucous membranes and a small cutaneous bite, resulting in neurotoxicity. To our knowledge, this is the first reported case of systemic neurotoxicity associated with ocular contact with coral snake venom. Our patient developed rapid-onset skeletal muscle weakness, which is very uncommon for M. tener, along with cranial nerve deficits. Acquisition of antivenom was challenging, but our patient provides a rare report of resolution of suspected M. tener neurotoxicity after receiving Central American coral snake (Micrurus nigrocinctus) antivenom. Our patient subsequently developed serum sickness, a known delayed complication of antivenom. WHY SHOULD AN EMERGENCY PHYSICIAN BE AWARE OF THIS?: The emergency physician should be aware that coral snake venom may be absorbed through different routes. Neurotoxicity and respiratory insufficiency may be fatal and onset may be delayed up to 13 h. North American Coral Snake Antivenom is in very limited supply, so non-Food and Drug Administration-approved alternative coral snake antivenoms may be used for patients demonstrating neurotoxicity. Emergency physicians should be proactive in contacting a toxicologist to procure antivenom, as well as consideration of adjunctive treatments, such as neostigmine. Furthermore, whole immunoglobulin G products, such as antivenom, may result in immediate and delayed reactions.
Subject(s)
Antivenins/pharmacology , Coral Snakes , Neurotoxicity Syndromes/drug therapy , Snake Venoms/adverse effects , Animals , Antivenins/therapeutic use , Female , Humans , Ocular Absorption , Poison Control Centers/organization & administration , Serum Sickness/etiology , Snake Bites/drug therapy , Snake Venoms/pharmacology , Thumb/injuries , Young AdultABSTRACT
Snakebite envenomation is a life-threatening disease that was recently re-included as a neglected tropical disease (NTD), affecting millions of people in tropical and subtropical areas of the world. Improvement in the therapeutic approaches to envenomation is required to palliate the morbidity and mortality effects of this NTD. The specific therapeutic treatment for this NTD uses snake antivenom immunoglobulins. Unfortunately, access to these vital drugs is limited, principally due to their cost. Different ethnic groups in the affected regions have achieved notable success in treatment for centuries using natural sources, especially plants, to mitigate the effects of snake envenomation. The ethnopharmacological approach is essential to identify the potential metabolites or derivatives needed to treat this important NTD. Here, the authors describe specific therapeutic snakebite envenomation treatments and conduct a review on different strategies to identify the potential agents that can mitigate the effects of the venoms. The study also covers an increased number of literature reports on the ability of natural sources, particularly plants, to treat snakebites, along with their mechanisms, drawbacks and future perspectives.
Subject(s)
Antivenins/therapeutic use , Snake Bites/drug therapy , Snake Venoms/adverse effects , Animals , Ethnopharmacology , Humans , Snake Bites/pathology , SnakesABSTRACT
Newly diagnosed glioblastoma multiforme with unmethylated MGMT promoter has a poor prognosis, with a median survival of 12 months. This phase II study investigated the efficacy and safety of combining the selective integrin inhibitor cilengitide with a combination of metronomic temozolomide and procarbazine for these patients. Eligible patients (newly diagnosed, histologically confirmed supratentorial glioblastoma with unmethylated MGMT promoter) were entered into this multicentre study. Cilengitide (2000 mg IV twice weekly) was commenced 1 week prior to radiotherapy combined with daily temozolomide (60 mg/m(2)) and procarbazine (50 or 100 mg) and, after 4 weeks' break, followed by six adjuvant cycles of temozolomide (50-60 mg/m(2)) and procarbazine (50 or 100 mg) on days 1-20, every 28 days. Cilengitide was continued for up to 12 months or until disease progression or unacceptable toxicity. The primary endpoint for efficacy was a 12-month overall survival rate of 65 %. Twenty-nine patients completed study treatment. Sixteen patients survived for 12 months or more, an overall survival rate of 55 %. The median overall survival was 14.5 months (95 % CI 11.1-19.6) and the median progression-free survival was 7.4 months (95 % CI 6.1-8). Cilengitide combined with metronomic temozolomide and procarbazine in MGMT-promoter unmethylated glioblastoma did not improve survival compared with historical data and does not warrant further investigation.
Subject(s)
Antineoplastic Agents/therapeutic use , Central Nervous System Neoplasms/therapy , Dacarbazine/analogs & derivatives , Glioblastoma/therapy , Procarbazine/therapeutic use , Snake Venoms/therapeutic use , Adult , Aged , Antineoplastic Agents/adverse effects , Central Nervous System Neoplasms/genetics , Chemoradiotherapy/adverse effects , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/adverse effects , Dacarbazine/therapeutic use , Disease Progression , Disease-Free Survival , Drug Therapy, Combination , Female , Glioblastoma/genetics , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Procarbazine/adverse effects , Snake Venoms/adverse effects , Temozolomide , Treatment Failure , Tumor Suppressor Proteins/genetics , Young AdultABSTRACT
BACKGROUND: This report describes a patient who developed coagulopathy after ingesting snake wine, which is an alcoholic libation containing an entire venomous snake. CASE REPORT: A 68-year-old man was admitted to the hospital 19 h after ingesting snake wine. The laboratory features upon admission included unmeasurable activated partial thromboplastin (aPTT) values, prolonged prothrombin time (PT) values, increased fibrinogen levels, modestly elevated fibrin degradation product and D-dimer values, uncorrected aPTT and PT values after a mixing test, and normal levels of aspartate transaminase and alanine transaminase. No pesticides, warfarin, or superwarfarin in the patient's blood or urine were detected. His coagulation profile normalized on the 6(th) day after admission after antivenom treatment. He was discharged 10 days later without sequelae. WHY SHOULD AN EMERGENCY PHYSICIAN BE AWARE OF THIS?: The physician should be aware that ingesting snake wine may lead to systemic envenomation. As for coagulopathy, which may develop by ingesting snake venom, related laboratory findings may differ from the features observed after direct envenomation by snakebite.
Subject(s)
Blood Coagulation Disorders/physiopathology , Serum Sickness/physiopathology , Snakes , Wine/adverse effects , Aged , Animals , Antivenins/pharmacology , Antivenins/therapeutic use , Blood Coagulation Disorders/blood , Blood Coagulation Tests , Humans , Male , Serum Sickness/therapy , Snake Venoms/adverse effectsABSTRACT
BACKGROUND: Recurrent and/or metastatic squamous cell carcinoma of the head and neck (R/M-SCCHN) overexpresses αvß5 integrin. Cilengitide selectively inhibits αvß3 and αvß5 integrins and is investigated as a treatment strategy. PATIENTS AND METHODS: The phase I/II study ADVANTAGE evaluated cilengitide combined with cisplatin, 5-fluorouracil, and cetuximab (PFE) in R/M-SCCHN. The phase II part reported here was an open-label, randomized, controlled trial investigating progression-free survival (PFS). Patients received up to six cycles of PFE alone or combined with cilengitide 2000 mg once (CIL1W) or twice (CIL2W) weekly. Thereafter, patients received maintenance therapy (cilengitide arms: cilengitide plus cetuximab; PFE-alone arm: cetuximab only) until disease progression or unacceptable toxicity. RESULTS: One hundred and eighty-two patients were treated. Median PFS per investigator read was similar for CIL1W + PFE, CIL2W + PFE, and PFE alone (6.4, 5.6, and 5.7 months, respectively). Accordingly, median overall survival and objective response rates were not improved with cilengitide (12.4 months/47%, 10.6 months/27%, and 11.6 months/36%, respectively). No clinically meaningful safety differences were observed between groups. None of the tested biomarkers (expression of integrins, CD31, Ki-67, vascular endothelial growth factor receptor 2, vascular endothelial-cadherin, type IV collagen, epidermal growth factor receptor, or p16 for human papillomavirus) were predictive of outcome. CONCLUSION: Neither of the cilengitide-containing regimens demonstrated a PFS benefit over PFE alone in R/M-SCCHN patients.
Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Cisplatin/therapeutic use , Fluorouracil/therapeutic use , Head and Neck Neoplasms/drug therapy , Snake Venoms/therapeutic use , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Cetuximab , Cisplatin/adverse effects , Disease Progression , Disease-Free Survival , ErbB Receptors/antagonists & inhibitors , Female , Fluorouracil/adverse effects , Humans , Male , Middle Aged , Snake Venoms/adverse effects , Squamous Cell Carcinoma of Head and Neck , Treatment OutcomeSubject(s)
Anti-Bacterial Agents/therapeutic use , Emergency Nursing/methods , Endophthalmitis/chemically induced , Endophthalmitis/drug therapy , Snake Venoms/adverse effects , Animals , Ciprofloxacin/therapeutic use , Endophthalmitis/nursing , Humans , Male , Middle Aged , Mydriatics/therapeutic use , Saline Solution , Tetracycline/therapeutic useABSTRACT
INTRODUCTION: This multicenter, open-label, phase II study was carried out to compare the efficacy and safety of cilengitide (EMD 121974), a selective inhibitor of the cell-surface integrins αVß3 and αVß5, with that of docetaxel in patients with advanced non-small-cell lung cancer (NSCLC). METHODS: Patients (n = 140) with advanced NSCLC who had failed first-line chemotherapy were randomized to cilengitide 240, 400, or 600 mg/m(2) twice weekly, or docetaxel 75 mg/m(2) once every 3 weeks for eight cycles. Non-progressing patients could continue cilengitide for up to 1 year. The primary endpoint was progression-free survival (PFS). No statistical tests were performed since the study was exploratory in nature and the number of patients enrolled was relatively small. RESULTS: Median PFS was 54, 63, 63, and 67 days for cilengitide 240, 400, and 600 mg/m(2), and docetaxel 75 mg/m(2), respectively. One-year survival rates were 13 %, 13 %, 29 %, and 27 %, respectively. The response rate (partial response only) with docetaxel was 15 %. No responses were reported in any cilengitide arm. The most frequent grade 3/4 treatment-related adverse events in the docetaxel group were leukopenia and neutropenia (experienced by 13 % of patients). Hematologic toxicity of this severity did not occur in cilengitide-treated patients. CONCLUSION: With the highest dose of cilengitide (600 mg/m(2)), median PFS and 1-year survival were similar to those in patients treated with docetaxel 75 mg/m(2) and there were fewer grade 3/4 treatment-related adverse events.
Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Integrins/antagonists & inhibitors , Lung Neoplasms/drug therapy , Snake Venoms/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Disease-Free Survival , Docetaxel , Female , Humans , Male , Middle Aged , Snake Venoms/adverse effects , Taxoids/administration & dosage , Taxoids/adverse effectsABSTRACT
BACKGROUND: Cilengitide (EMD121974) is a cyclized pentapeptide that is a potent and selective integrin antagonist which has shown activity in malignant gliomas. In all previous studies, cilengitide has been administered in an intermittent fashion. However, cilengitide has a short half-life of 3-5 h with no evidence of drug accumulation. These data prompted the initiation of this phase I study of continuous infusion cilengitide. METHODS: Cilengitide was administered as a continuous infusion without break in 4-week cycles. Plasma samples for pharmacokinetic studies were obtained weekly in cycle 1 immediately prior to and 2 h after infusion bag change. RESULTS: Thirty-five patients were treated (median age 56; 23 males) at dose levels of 1, 2, 4, 8, 12, 18, 27, and 40 mg/h. Toxicities were limited to grade ≤ 2 and showed no relation to dose. Fatigue was most common (17%), while all other toxicities were reported in <10% of patients. No dose-limiting toxicities were observed, and therefore the maximum tolerated dose was not reached. Pharmacokinetic analysis showed that values for clearance and volume of distribution were comparable across dose levels, and the steady-state concentration increased proportionally with dose. CONCLUSIONS: Cilengitide can be safely administered as a continuous infusion at doses up to at least 40 mg/h, which represents the maximum feasible dose due to drug solubility and delivery limitations. The pharmacokinetics of continuous infusion cilengitide are linear and consistent with the results obtained using a twice weekly infusion.
Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Snake Venoms/administration & dosage , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Chicago , Female , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/blood , Neoplasms/pathology , Snake Venoms/adverse effects , Snake Venoms/blood , Snake Venoms/pharmacokinetics , Treatment OutcomeABSTRACT
BACKGROUND: Integrins mediate invasion and angiogenesis in prostate cancer bone metastases. We conducted a phase II study of cilengitide, a selective antagonist of α(v)ß(3) and α(v)ß(5) integrins, in non-metastatic castration resistant prostate cancer with rising PSA. METHODS: Patients were observed for 4 weeks with PSA monitoring, and then treated with 2,000 mg IV of cilengitide twice weekly until toxicity/progression. PSA, circulating tumor cells (CTCs) and circulating endothelial cells (CECs) were monitored each cycle with imaging performed every three cycles. Primary end point was PSA decline by ≥ 50%. Secondary endpoints were safety, PSA slope, time to progression (TTP), overall survival (OS), CTCs, CECs and gene expression. RESULTS: 16 pts were enrolled; 13 were eligible with median age 65.5 years, baseline PSA 8.4 ng/mL and median Gleason sum 7. Median of three cycles was administered. Treatment was well tolerated with two grade three toxicities and no grade four toxicities. There were no PSA responses; 11 patients progressed by PSA after three cycles. Median TTP was 1.8 months and median OS has not been reached. Median pre- and on-treatment PSA slopes were 1.1 and 1.8 ng/mL/month. Baseline CTCs were detected in 1/9 patients. CTC increased (0 to 1; 2 pts), remained at 0 (2 pts) or decreased (23 to 0; 1 patient) at progression. Baseline median CEC was 26 (0-61) and at progression, 47 (15-148). Low cell counts precluded gene expression studies. CONCLUSIONS: Cilengitide was well tolerated but had no detectable clinical activity. CTCs are of questionable utility in non-metastatic prostate cancer.
Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Prostatic Neoplasms/drug therapy , Snake Venoms/therapeutic use , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Disease Progression , Drug Administration Schedule , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Infusions, Intravenous , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/metabolism , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Grading , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/pathology , Prostate-Specific Antigen/blood , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Receptors, Vitronectin/antagonists & inhibitors , Receptors, Vitronectin/metabolism , Snake Venoms/administration & dosage , Snake Venoms/adverse effects , Time Factors , Treatment Failure , United StatesABSTRACT
Cilengitide is a cyclic pentapeptide that is a specific inhibitor of the αvß3 and αvß5 integrins. Preclinical studies demonstrate antiangiogenic activity and anti-invasive activity in a number of glioma models. This study was designed to evaluate the efficacy and tumor delivery of cilengitide in patients with recurrent glioblastoma. Patients with recurrent glioblastoma who require a surgical resection for optimal clinical care received 3 intravenous doses of cilengitide at either 500 or 2000 mg (day -8, -4, -1) prior to undergoing tumor resection with corresponding blood samples for plasma to tumor comparisons. After recovery from surgery, patients were treated with cilengitide (2000 mg i.v. twice weekly, maximum of 2 years of treatment). The study accrued 30 patients with recurrent glioblastoma, 26 were evaluable for efficacy. The 6-month progression free survival rate was 12%. Cilengitide was detected in all tumor specimens with higher levels in the group receiving 2000 mg dosing while corresponding plasma concentrations were low, often below the lower limit of detection. These results confirm drug delivery and possibly retention in tumor. This study provides evidence that with established dosing, cilengitide is adequately delivered to the tumor, although as a single agent, efficacy in recurrent glioblastoma is modest. However, these results demonstrating drug delivery to tumor do support continued investigation of this agent as preliminary results from recent studies combining cilengitide with cytotoxic therapies are promising.
Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Snake Venoms/therapeutic use , Adult , Aged , Brain Neoplasms/surgery , Combined Modality Therapy , Disease-Free Survival , Endpoint Determination , Female , Glioblastoma/surgery , Humans , Immunotherapy , Integrins/physiology , Karnofsky Performance Status , Male , Middle Aged , Recurrence , Snake Venoms/adverse effects , Snake Venoms/pharmacokineticsSubject(s)
Snake Bites/epidemiology , Snake Bites/therapy , Wilderness Medicine/standards , Animals , Antivenins/administration & dosage , Antivenins/therapeutic use , Humans , Missouri , Snake Bites/mortality , Snake Venoms/adverse effects , Snake Venoms/metabolism , Species Specificity , United States/epidemiologyABSTRACT
BACKGROUND: Integrins are involved in prostate cancer metastasis by regulating cell adhesion, migration, invasion, motility, angiogenesis and bone metabolism. We evaluated the efficacy of two dose levels of cilengitide in patients (pts) with castrate resistant prostate cancer (CRPC). METHODS: Chemotherapy-naïve, asymptomatic metastatic CRPC pts were randomized to cilengitide 500 mg or 2,000 mg IV twice weekly using parallel 2-stage design. The primary endpoint was rate of objective clinical progression at 6-months. Secondary endpoints included clinical and PSA response rates, safety and effects of cilengitide treatment on circulating tumor cells (CTCs) and bone remodeling markers. RESULTS: Forty-four pts were accrued to first stage (22/arm). Median number of cycles was three in both arms (500 mg arm: 1-8; 2,000 mg arm: 1-15). At 6 months, two pts (9%) on the 500 mg arm and five pts (23%) on the 2,000 mg arm had not progressed. Best objective response was stable disease (SD) in seven pts for 9.9[8.1,20.9] months. There were three grade 3 and no grade 4 toxicities. At 12 weeks, analysis of bone markers did not reveal significant trends. At progression, bone specific alkaline phosphatase and N-telopeptide increased in all pts, less so in pts on the 2,000 mg arm and in pts on both arms who obtained SD at 6 months. CTCs increased over time in both arms. CONCLUSION: Cilengitide was well tolerated with modest clinical effect in favor of the higher dose. The unique trial design including a shift from response rate to objective progression as the endpoint, and not acting on PSA increases was feasible.
Subject(s)
Antineoplastic Agents/therapeutic use , Prostate-Specific Antigen/drug effects , Prostatic Neoplasms/drug therapy , Snake Venoms/therapeutic use , Aged , Aged, 80 and over , Alkaline Phosphatase/metabolism , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Collagen Type I/metabolism , Disease Progression , Dose-Response Relationship, Drug , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplastic Cells, Circulating/drug effects , Peptides/metabolism , Prostatic Neoplasms/pathology , Snake Venoms/administration & dosage , Snake Venoms/adverse effects , Time Factors , Treatment OutcomeABSTRACT
Cilengitide, a cyclicized arginine-glycine-aspartic acid-containing pentapeptide, potently blocks ανß3 and ανß5 integrin activation. Integrins are upregulated in many malignancies and mediate a wide variety of tumor-stroma interactions. Cilengitide and other integrin-targeting therapeutics have preclinical activity against many cancer subtypes including glioblastoma (GBM), the most common and deadliest CNS tumor. Cilengitide is active against orthotopic GBM xenografts and can augment radiotherapy and chemotherapy in these models. In Phase I and II GBM trials, cilengitide and the combination of cilengitide with standard temozolomide and radiation demonstrate consistent antitumor activity and a favorable safety profile. Cilengitide is currently under evaluation in a pivotal, randomized Phase III study (Cilengitide in Combination With Temozolomide and Radiotherapy in Newly Diagnosed Glioblastoma Phase III Randomized Clinical Trial [CENTRIC]) for newly diagnosed GBM. In addition, randomized controlled Phase II studies with cilengitide are ongoing for non-small-cell lung cancer and squamous cell carcinoma of the head and neck. Cilengitide is the first integrin inhibitor in clinical Phase III development for oncology.
Subject(s)
Antineoplastic Agents/therapeutic use , Glioblastoma/drug therapy , Integrins/antagonists & inhibitors , Oligopeptides/therapeutic use , Snake Venoms/therapeutic use , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Clinical Trials as Topic , Drug Evaluation, Preclinical , Drug Industry/trends , Humans , Oligopeptides/adverse effects , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Snake Venoms/adverse effects , Snake Venoms/chemistry , Snake Venoms/pharmacokinetics , Treatment OutcomeABSTRACT
Animal venoms, widespread throughout the world, are complex mixtures, the composition of which depends on the venom-producing species. The objective of this study was to contribute to the development of animal venom-based medicines by investigating the use of animal venom pharmacopuncture in Korean medicine (KM) institutions. We surveyed 256 public health centers from 1 through 31 October 2019 as guided by the Ministry of Health and Welfare (MoHW). A questionnaire developed by an expert group was distributed and collected for statistical analysis. The survey identified three types of animal venom-based pharmacopuncture: bee, snake, and toad venoms. The medications are based on a single animal venom ingredient and produced in 11 external herbal dispensaries (EHDs). Each animal venom is processed, refined, and freeze-dried in a cleanroom to produce a powder formulation that is later measured, diluted, filtered, filled, sealed, sterilized, and packaged as pharmacopuncture injections used in KM institutions. Bee venom therapy is effective in treating musculoskeletal pain, snake venom therapy is effective in controlling bleeding during surgery, and toad venom therapy is effective in cancer treatment. The study suggests that bee, snake, and toad venoms could be used in medical institutions and have the potential for drug development.
Subject(s)
Acupuncture Therapy , Amphibian Venoms/therapeutic use , Bee Venoms/therapeutic use , Medicine, Korean Traditional , Snake Venoms/therapeutic use , Acupuncture Therapy/adverse effects , Amphibian Venoms/adverse effects , Animals , Bee Venoms/adverse effects , Humans , Republic of Korea , Snake Venoms/adverse effects , Treatment OutcomeABSTRACT
The methanolic extract of fresh tea leaves of Camellia sinensis L. (Theaceae) (CS) was assayed for its potential to inhibit enzymes with hydrolytic activity in Naja naja kaouthia Lesson (Elapidae) and Calloselasma rhodostoma Kuhl (Viperidae) venoms. These snake venom enzymes are responsible for the early effects of envenomation, such as local tissue damage and inflammation. The CS extract inhibited phospholipase A(2), proteases, hyaluronidase and L-amino acid oxidase in both venoms by in vitro neutralization and inhibited the hemorrhagic and the dermonecrotic activities of the venoms in vivo. It is suggested that the inhibitory potential of the CS extract against local tissue damage induced by snake venoms may be attributed to complexation and chelation between the venom proteins and the phenolic contents of the extract.
Subject(s)
Antivenins/pharmacology , Flavonoids/pharmacology , Phenols/pharmacology , Plant Extracts/pharmacology , Snake Venoms/adverse effects , Tea/chemistry , Animals , Elapidae , Hyaluronoglucosaminidase/antagonists & inhibitors , L-Amino Acid Oxidase/antagonists & inhibitors , Male , Mice , Necrosis/drug therapy , Necrosis/prevention & control , Phospholipases A/antagonists & inhibitors , Polyphenols , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Snake Bites/drug therapy , Snake Venoms/enzymology , ViperidaeABSTRACT
The application of angiogenesis inhibitors in neurooncology is increasing. Initially, these drugs seemed to be very promising because of the surprisingly high neuroradiological response rates that were observed in first clinical trials. Meanwhile, this enthusiasm is waning, as the high response rates did not translate into substantial improvements in progression-free and overall survival. Tumor progression during or after antiangiogenic therapy is often associated with rapid clinical neurological deterioration and sometimes even with diffuse infiltrative gliomatosis-like neuroradiological phenotypes. Thus, the characterization and understanding of escape mechanisms are needed. The identification of criteria for defining the personalized use of angiogenesis inhibitors remains a challenge.
Subject(s)
Angiogenesis Inhibitors/therapeutic use , Brain Neoplasms/blood supply , Brain Neoplasms/drug therapy , Glioma/blood supply , Glioma/drug therapy , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Bevacizumab , Brain Neoplasms/mortality , Clinical Trials as Topic , Disease-Free Survival , Drug Resistance, Neoplasm , Glioma/mortality , Humans , Microcirculation/drug effects , Neoplasm Recurrence, Local/blood supply , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/mortality , Prognosis , Quinazolines/adverse effects , Quinazolines/therapeutic use , Snake Venoms/adverse effects , Snake Venoms/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitorsABSTRACT
Snakebite envenoming is a neglected, public health problem in tropical and subtropical regions. Local tissue necrosis, neurotoxic, and hemo-vasculotoxic effects are well-recognized features, whereas the endocrine and metabolic derangements are not as well known. In addition to contributing to morbidity, some of these manifestations can be potentially life-threatening if not recognized early. The most prominent endocrine manifestation is hypopituitarism (HP), which can manifest acutely or remain asymptomatic and present years later. Unexplained recurrent hypoglycemia and refractory hypotension are early clinical clues to suspect corticotroph axis involvement in acute settings. Chronic pituitary failure may present, like Sheehan's syndrome, several years after the bite. The occurrence of acute kidney injury, capillary leak syndrome, and disseminated intravascular coagulation are predictors of HP. Adrenal hemorrhages are documented in autopsy series; however, primary adrenal insufficiency is very rare and confounded by the presence of HP. Hyponatremia, hypokalemia or hyperkalemia, and dysglycemia can occur, but the mechanisms involved are only partially understood. Awareness, a high index of suspicion, correct interpretation of hormonal parameters, and timely treatment of these abnormalities can be lifesaving.
Subject(s)
Hypopituitarism , Snake Bites , Snake Venoms/adverse effects , Adrenal Insufficiency , Humans , Hypoglycemia , Hyponatremia , Hypopituitarism/etiology , Hypopituitarism/pathology , Pituitary Gland/physiopathology , Snake Bites/pathology , Snake Bites/therapyABSTRACT
Snakebite envenoming (SBE) is a public health issue in developing countries. The estimated annual global incidence of snakebites is about 5.4 million snakebites per year, resulting from 1.8 to 2.7 million cases of SBE and from 81,000 to 138,000 deaths with 400,000 survivors suffering permanent physical and psychological disabilities. There are more than 3000 species of snakes around the world: 600 are venomous and over 200 are considered to be medically important because of their clinical effects. The severity of SBE depends on several factors among which bite localization, snake's size, condition of glands and teeth, bite angle and bite duration, the microflora of the snake's mouth and victim's skin, age of the victim, weight, health status, and victim's activity after a bite. Snake venoms are mixtures of protein families, and each of these families contains many different toxins or toxin isoforms. Based on their effects, snake venoms can be classified as hemotoxic, neurotoxic, or cytotoxic and they can all act together involving multiple tissues and organs. When the bite is fatal, the mechanism of death is primarily related to the paralysis of respiratory muscles, which causes asphyxia and hypoxic-ischemic encephalopathy, but also anaphylactic shock, hemorrhagic shock, cardiomyopathy, acute tubular necrosis (ATN). The purpose of this literature review is to evaluate epidemiological and post-mortem examination findings in fatal SBEs in order to better understand the pathophysiological mechanisms, thus helping pathologists in defining the correct diagnosis.