Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Ethnopharmacol ; 316: 116734, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37290735

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Processed from natural minerals, arsenic trioxide (ATO) as an ancient Chinese medicine has been used to treat diseases for over 2000 years. And it was applied to treat acute promyelocytic leukemia (APL) since the 1970s in China. Summarizing the clinical evidence of ATO in cancer is conducive to further understanding, development, and promotion of its pharmacological research. AIM OF THE STUDY: It is the first time to comprehensively assess and summarize the evidence of ATO in cancer treatment via umbrella review. MATERIALS AND METHODS: 8 databases in English or Chinese from their inception to February 21, 2023 were searched by two reviewers separately and suitable meta-analyses (MAs) were included in this umbrella review. Their methodological quality and risk of bias were evaluated and data of outcomes was extracted and pooled again. The evidence certainty of pooled results was classified. RESULTS: 17 MAs with 27 outcomes and seven comparisons in three cancers were included in this umbrella review. However, their methodological quality was unsatisfactory with 6 MAs as low quality and 12 MAs as critically low quality. Their shortcomings were mainly focused on protocol, literature selecting, bias risk, small sample study bias, and conflicts of interest or funding. And they were all assessed as high risk in bias. It was suggested that ATO had an advantage in enhancing complete remission rate, event-free survival, and recurrence free survival and decreasing recurrence rate, cutaneous toxicity, hyper leukocyte syndrome, tretinoin syndrome, edema and hepatotoxicity in different comparisons of APL with low or moderate certainty. Besides, compared with transcatheter arterial chemoembolization (TACE) alone, ATO plus TACE also could improve objective response rate, disease control rate, survival rate (0.5, 1, 2, and 3-year) and life quality and reduce the level of alpha fetoprotein in primarily hepatocellular carcinoma with low or moderate certainty. However, no significant results were found in MM. Finally, key findings were as followed. ATO has potential broad-spectrum anticancer effects but the clinical transformation is rarely achieved. Route of administration may affect the antitumor effects of ATO. ATO can act synergistically in combination with a variety of antitumor therapies. The safety and drug resistance of ATO should be paid more attention to. CONCLUSIONS: ATO may be a promising drug in anticancer treatment although earlier RCTs have dragged down the level of evidence. However, high-quality clinical trials are expected to explore its broad-spectrum anticancer effects, wide application, appropriate route of administration, and compound dosage form.


Subject(s)
Carcinoma, Hepatocellular , Chemoembolization, Therapeutic , Leukemia, Promyelocytic, Acute , Liver Neoplasms , Humans , Arsenic Trioxide/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Randomized Controlled Trials as Topic , Leukemia, Promyelocytic, Acute/drug therapy , Tretinoin/therapeutic use
2.
Acta Pharmacol Sin ; 44(4): 822-831, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36216898

ABSTRACT

The acute promyelocytic leukemia (APL) driver ZBTB16/RARα is generated by the t(11;17) (q23;q21) chromosomal translocation, which is resistant to combined treatment of all-trans retinoic acid (ATRA) and arsenic trioxide (ATO) or conventional chemotherapy, resulting in extremely low survival rates. In the current study, we investigated the effects of hyperthermia on the oncogenic fusion ZBTB16/RARα protein to explore a potential therapeutic approach for this variant APL. We showed that Z/R fusion protein expressed in HeLa cells was resistant to ATO, ATRA, and conventional chemotherapeutic agents. However, mild hyperthermia (42 °C) rapidly destabilized the ZBTB16/RARα fusion protein expressed in HeLa, 293T, and OCI-AML3 cells, followed by robust ubiquitination and proteasomal degradation. In contrast, hyperthermia did not affect the normal (i.e., unfused) ZBTB16 and RARα proteins, suggesting a specific thermal sensitivity of the ZBTB16/RARα fusion protein. Importantly, we found that the destabilization of ZBTB16/RARα was the initial step for oncogenic fusion protein degradation by hyperthermia, which could be blocked by deletion of nuclear receptor corepressor (NCoR) binding sites or knockdown of NCoRs. Furthermore, SIAH2 was identified as the E3 ligase participating in hyperthermia-induced ubiquitination of ZBTB16/RARα. In short, these results demonstrate that hyperthermia could effectively destabilize and subsequently degrade the ZBTB16/RARα fusion protein in an NCoR-dependent manner, suggesting a thermal-based therapeutic strategy that may improve the outcome in refractory ZBTB16/RARα-driven APL patients in the clinic.


Subject(s)
Hyperthermia, Induced , Leukemia, Promyelocytic, Acute , Humans , Antineoplastic Agents/pharmacology , Arsenic Trioxide/therapeutic use , HeLa Cells , Leukemia, Promyelocytic, Acute/therapy , Leukemia, Promyelocytic, Acute/drug therapy , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Oncogene Proteins, Fusion/therapeutic use , Promyelocytic Leukemia Zinc Finger Protein/genetics , Tretinoin/pharmacology , Tretinoin/therapeutic use
3.
Cells ; 11(21)2022 11 01.
Article in English | MEDLINE | ID: mdl-36359850

ABSTRACT

The turn-on mutations of the KRAS gene, coding a small GTPase coupling growth factor signaling, are contributing to nearly 25% of all human cancers, leading to highly malignant tumors with poor outcomes. Targeting of oncogenic KRAS remains a most challenging task in oncology. Recently, the specific G12C mutant KRAS inhibitors have been developed but with a limited clinical outcome because they acquire drug resistance. Alternatively, exploiting a metabolic breach of KRAS-mutant cancer cells related to a glucose-dependent sensitivity to oxidative stress is becoming a promising indirect cancer targeting approach. Here, we discuss the use of a vitamin C (VC) acting in high dose as an oxidative "Trojan horse" agent for KRAS-mutant cancer cells that can be potentiated with another oxidizing drug arsenic trioxide (ATO) to obtain a potent and selective cytotoxic impact. Moreover, we outline the advantages of VC's non-natural enantiomer, D-VC, because of its distinctive pharmacokinetics and lower toxicity. Thus, the D-VC and ATO combination shows a promising path to treat KRAS-mutant cancers in clinical settings.


Subject(s)
Ascorbic Acid , Neoplasms , Humans , Arsenic Trioxide/pharmacology , Arsenic Trioxide/therapeutic use , Ascorbic Acid/pharmacology , Ascorbic Acid/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Oxidative Stress , Vitamins/pharmacology , Oxidation-Reduction , Neoplasms/drug therapy , Neoplasms/genetics
4.
J Control Release ; 350: 761-776, 2022 10.
Article in English | MEDLINE | ID: mdl-36063961

ABSTRACT

Arsenotherapy has been clinically exploited to treat a few types of solid tumors despite of acute promyelocytic leukemia using arsenic trioxide (ATO), however, its efficacy is hampered by inadequate delivery of ATO into solid tumors owing to the absence of efficient and biodegradable vehicles. Precise spatiotemporal control of subcellular ATO delivery for potent arsenotherapy thus remains challengeable. Herein, we report the self-activated arsenic manganite nanohybrids for high-contrast magnetic resonance imaging (MRI) and arsenotherapeutic synergy on triple-negative breast cancer (TNBC). The nanohybrids, composed of arsenic­manganese-co-biomineralized nanoparticles inside albumin nanocages (As/Mn-NHs), switch signal-silent background to high proton relaxivity, and simultaneously afford remarkable subcellular ATO level in acidic and glutathione environments, together with reduced ATO resistance against tumor cells. Then, the nanohybrids enable in vivo high-contrast T1-weighted MRI signals in various tumor models for delineating tumor boundary, and simultaneously yield efficient arsenotherapeutic efficacy through multiple apoptotic pathways for potently suppressing subcutaneous and orthotopic breast models. As/Mn-NHs exhibited the maximum tumor-to-normal tissue (T/N) contrast ratio of 205% and tumor growth inhibition rate of 88% at subcutaneous 4T1 tumors. These nanohybrids further yield preferable synergistic antitumor efficacy against both primary and metastatic breast tumors upon combination with concurrent thermotherapy. More importantly, As/Mn-NHs considerably induce immunogenic cell death (ICD) effect to activate the immunogenically "cold" tumor microenvironment into "hot" one, thus synergizing with immune checkpoint blockade to yield the strongest tumor inhibition and negligible metastatic foci in the lung. Our study offers the insight into clinically potential arsenotherapeutic nanomedicine for potent therapy against solid tumors.


Subject(s)
Antineoplastic Agents , Arsenic , Arsenicals , Neoplasms , Albumins , Apoptosis , Arsenic/pharmacology , Arsenic/therapeutic use , Arsenic Trioxide/pharmacology , Arsenic Trioxide/therapeutic use , Arsenicals/therapeutic use , Cell Line, Tumor , Glutathione/pharmacology , Humans , Immune Checkpoint Inhibitors , Manganese , Manganese Compounds , Neoplasms/drug therapy , Oxides , Protons , Tumor Microenvironment
5.
J Immunol Res ; 2022: 2619781, 2022.
Article in English | MEDLINE | ID: mdl-35178457

ABSTRACT

Hepatocellular carcinoma (HCC) is an often-fatal malignant tumor with high lethality. Despite advances and significant efficacy in monotherapy, cancer therapy continues to pose several challenges. Novel combination regimens are an emerging strategy for anti-HCC and have demonstrated to be effective. Here, we propose a potential combination for HCC treatment named arsenic trioxide cooperate cryptotanshinone (ACCS). A remarkable synergistic therapeutic effect has been achieved compared with drugs alone in both in vivo and in vitro experiments. Mechanism study indicated that ACCS exerts its therapeutic actions by regulating macrophage-related immunity and glycolysis. ACCS potentiates the polarization of M1 macrophages and elevates the proportion of M1/M2 to remodel tumor immunity. Further molecular mechanism study revealed that ACCS intensifies the glucose utilization and glycolysis in the macrophage by increasing the phosphorylation of AMPK to activating the AMPK singling pathway. In conclusion, ACCS is a highly potential combination regimen for HCC treatment. The therapeutic potential of ACCS as a candidate option for anticancer drugs in restoring the balance of immunity and metabolism deserves further investigation.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenic Trioxide/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Drugs, Chinese Herbal/therapeutic use , Liver Neoplasms/drug therapy , Macrophages/metabolism , Phenanthrenes/therapeutic use , Animals , Cell Differentiation , Cytokines/metabolism , Drug Combinations , Drug Synergism , Glycolysis , Humans , Immunity, Innate , Immunomodulation , Macrophage Activation , Mice , Mice, Inbred BALB C , Th1 Cells/immunology
6.
Int J Pharm ; 609: 121209, 2021 Nov 20.
Article in English | MEDLINE | ID: mdl-34678398

ABSTRACT

Arsenic trioxide (ATO) is the active ingredient in traditional Chinese medicine, i.e., Arsenic, which has shown excellent therapeutic effects on hepatocellular carcinoma. However, due to its poor tumor distribution and high toxicity, the mass adoption of ATO in clinical applications has been severely impeded. In this study, matrix metalloproteinase 2 (MMP2)-responsive cleaved cell-penetrating peptide (PF) and folate (FA) co-modified liposome coated calcium arsenate nanoparticles (FA/PF-LP-CaAs) were fabricated based on these two considerations: (1) The tumor microenvironment characterized by overexpressed MMP2 in extracellular matrix and folate receptor on the cell membrane can enhance drug accumulation and accelerate endocytosis; (2) leveraging different toxicity of arsenic in different valence states, i.e., AsV can be reduced to more toxic AsIII by glutathione in tumor cells. Furthermore, FA/PF-LP-CaAs could be responsively degraded by the mild acidic tumor environment, and the degraded product could escape from lysosomes after endocytosis. More importantly, in light of the in vivo biodistribution and pharmacodynamic studies, the vehicle was able to accumulate in the tumor efficiently. Also, it was able to exhibit excellent anti-tumor efficacy with minimized side effects when compared to single-modified counterparts. Thus, the novel strategy based on the tumor microenvironment proposed in this work can enhance the tumor-targeting efficiency and intratumor toxicity.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Prodrugs , Antineoplastic Agents/therapeutic use , Arsenic Trioxide/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Drug Delivery Systems , Humans , Liver Neoplasms/drug therapy , Matrix Metalloproteinase 2 , Prodrugs/therapeutic use , Tissue Distribution , Tumor Microenvironment
7.
Cancer ; 127(8): 1186-1207, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33734442

ABSTRACT

The unraveling of the pathophysiology of acute myeloid leukemia (AML) has resulted in rapid translation of the information into clinical practice. After more than 40 years of slow progress in AML research, the US Food and Drug Administration has approved nine agents for different AML treatment indications since 2017. In this review, we detail the progress that has been made in the research and treatment of AML, citing key publications related to AML research and therapy in the English literature since 2000. The notable subsets of AML include acute promyelocytic leukemia (APL), core-binding factor AML (CBF-AML), AML in younger patients fit for intensive chemotherapy, and AML in older/unfit patients (usually at the age cutoff of 60-70 years). We also consider within each subset whether the AML is primary or secondary (therapy-related, evolving from untreated or treated myelodysplastic syndrome or myeloproliferative neoplasm). In APL, therapy with all-trans retinoic acid and arsenic trioxide results in estimated 10-year survival rates of ≥80%. Treatment of CBF-AML with fludarabine, high-dose cytarabine, and gemtuzumab ozogamicin (GO) results in estimated 10-year survival rates of ≥75%. In younger/fit patients, the "3+7" regimen (3 days of daunorubicin + 7 days of cytarabine) produces less favorable results (estimated 5-year survival rates of 35%; worse in real-world experience); regimens that incorporate high-dose cytarabine, adenosine nucleoside analogs, and GO are producing better results. Adding venetoclax, FLT3, and IDH inhibitors into these regimens has resulted in encouraging preliminary data. In older/unfit patients, low-intensity therapy with hypomethylating agents (HMAs) and venetoclax is now the new standard of care. Better low-intensity regimens incorporating cladribine, low-dose cytarabine, and other targeted therapies (FLT3 and IDH inhibitors) are emerging. Maintenance therapy now has a definite role in the treatment of AML, and oral HMAs with potential treatment benefits are also available. In conclusion, AML therapy is evolving rapidly and treatment results are improving in all AML subsets as novel agents and strategies are incorporated into traditional AML chemotherapy. LAY SUMMARY: Ongoing research in acute myeloid leukemia (AML) is progressing rapidly. Since 2017, the US Food and Drug Administration has approved 10 drugs for different AML indications. This review updates the research and treatment pathways for AML.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Age Factors , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arsenic Trioxide/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cladribine/therapeutic use , Core Binding Factors , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Gemtuzumab/therapeutic use , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/mortality , Maintenance Chemotherapy , Mutation , Myelodysplastic Syndromes/complications , Myeloproliferative Disorders/complications , Neoplasm, Residual , Sulfonamides/therapeutic use , Survival Rate , Translational Research, Biomedical , Tretinoin/therapeutic use , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use
8.
Leuk Lymphoma ; 62(3): 703-708, 2021 03.
Article in English | MEDLINE | ID: mdl-33107373

ABSTRACT

Arsenic trioxide (ATO) is the backbone of acute promyelocytic leukemia (APL) treatment and is dosed based on weight with no upper limit, therefore obese patients receive large doses and may be vulnerable to adverse effects and dose holdings. Twenty-seven patients receiving ATO during induction were categorized as obese (N = 16) or non-obese (N = 11) based on body mass index (BMI) ≥30 kg/m2 in this retrospective study. Doses were held or modified due to composite adverse effects in 9 (56%) obese patients and 7 (64%) non-obese patients (p = 1.00). There were higher rates of dose holdings (13% versus 0%; p = .5) and dose modifications (13% versus 0%; p = .5) due to hepatotoxicity in obese versus non-obese patients. There were no differences in efficacy parameters. These data suggest that obese patients have similar overall incidence of adverse effects to ATO as non-obese patients; any difference in risk of hepatotoxicity will require clarification in a larger study.


Subject(s)
Arsenicals , Drug-Related Side Effects and Adverse Reactions , Leukemia, Promyelocytic, Acute , Antineoplastic Combined Chemotherapy Protocols , Arsenic Trioxide/therapeutic use , Arsenicals/adverse effects , Humans , Leukemia, Promyelocytic, Acute/complications , Leukemia, Promyelocytic, Acute/drug therapy , Obesity/complications , Oxides/adverse effects , Retrospective Studies , Tretinoin/therapeutic use
9.
Metallomics ; 12(3): 326-336, 2020 03 01.
Article in English | MEDLINE | ID: mdl-32163072

ABSTRACT

Arsenic trioxide (ATO) has been recognized as a drug for the treatment of various diseases in traditional medicine for more than two thousand years. Although ATO has recently shown excellent efficacy for the treatment of acute promyelocytic leukemia (APL), it could not provide satisfactory outcomes as a single-agent for the management of non-APL leukemia or different solid tumors. Nevertheless, combination treatment strategies, e.g., ATO with other agents, have shown promising results against different diseases. Here, we introduce in depth the latest evidence and detailed insights into ATO-mediated cures for APL by targeting PML/RARα chimeric protein, followed by the preclinical and clinical efficacy of ATO on various non-APL malignancies and solid tumors. Likewise, the antiviral activity of ATO against human immunodeficiency virus (HIV) and hepatitis C virus (HCV) was also discussed briefly. Our review would provide a clear prospect for the combination of ATO with other agents for treatment of numerous neoplastic diseases, and open a new era in the clinically applicable range of arsenicals.


Subject(s)
Antineoplastic Agents/therapeutic use , Antiviral Agents/therapeutic use , Arsenic Trioxide/therapeutic use , Neoplasms/drug therapy , Virus Diseases/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antiviral Agents/pharmacology , Arsenic Trioxide/pharmacology , Humans , Leukemia, Promyelocytic, Acute/drug therapy
10.
ACS Appl Mater Interfaces ; 12(7): 8016-8029, 2020 Feb 19.
Article in English | MEDLINE | ID: mdl-31997633

ABSTRACT

Arsenic trioxide (ATO) is effective in the treatment of hematological malignancies and solid tumors. However, its toxicity and side effects are severe, posing an obstacle in its clinical application. A controlled-release ATO carrier with mitochondrial targeting was constructed in this study. The safety and efficacy in vitro were investigated using a hemolysis test, cytotoxicity, proliferation, migration, apoptosis, and other changes in cell behavior. The safety and efficacy were further evaluated in vivo by hematoxylin-eosin staining, terminal deoxyribonucleotide transferase-mediated dUTP nick end labeling staining, and blood testing in tumor-bearing mice. Immunohistochemically and western blotting experiments were conducted to explore the mechanism of combination therapy of material-based chemotherapy and microwave hyperthermia in vitro. We demonstrated that the nano-zirconia (ZrO2) loading platform may be used to administer the ATO, with local precision-controlled release and mitochondrial targeting. Furthermore, we showed the safety of this approach for delivering high doses of ATO. In addition, we explored this new method in combination with in vitro microwave heat therapy, providing a potentially novel intravenous approach to chemotherapy. We described a new non-invasive treatment that improved the efficacy of ATO chemotherapy against hepatocellular carcinoma through nano-ZrO2 carriers.


Subject(s)
Antineoplastic Agents/administration & dosage , Arsenic Trioxide/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Drug Carriers/chemistry , Hyperthermia, Induced/methods , Liver Neoplasms/drug therapy , Mitochondria/drug effects , Nanoparticles/chemistry , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Arsenic Trioxide/pharmacology , Arsenic Trioxide/therapeutic use , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Movement/drug effects , Cell Proliferation/drug effects , Delayed-Action Preparations , Drug Liberation , Hep G2 Cells , Humans , Hyperthermia, Induced/instrumentation , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Nanoparticles/ultrastructure , Particle Size , Xenograft Model Antitumor Assays , Zirconium/chemistry
11.
Am J Hematol ; 93(12): 1467-1473, 2018 12.
Article in English | MEDLINE | ID: mdl-30160789

ABSTRACT

Intravenous arsenic trioxide (ATO) has been adopted as the first-line treatment for acute promyelocytic leukemia (APL). Another arsenic compound named the Realgar-Indigo naturalis formula (RIF), an oral traditional Chinese medicine containing As4 S4 , has been shown to be highly effective in treating adult APL. In the treatment of pediatric APL, the safety and efficacy of RIF remains to be confirmed. This randomized, multicenter, and noninferiority trial was conducted to determine whether intravenous ATO can be substituted by oral RIF in the treatment of pediatric APL. From September 2011 to January 2017, among 92 patients who were 16 years old or younger with newly diagnosed PML-RARa positive APL, 82 met eligible criteria and were randomly assigned to ATO (n = 42) or RIF (n = 40) group. The remaining 10 patients did not fulfilled eligible criteria because five did not accept randomization, four died and one had hemiplegia prior to arsenic randomization due to intracranial hemorrhage or cerebral thrombosis. Induction and consolidation treatment contained ATO or RIF, all-trans-retinoic acid and low intensity chemotherapy. End points included event-free survival (EFS), adverse events and hospital days. After a median 3-year follow-up, the estimated 5-year EFS was 100% in both groups, and adverse events were mild. However, patients in the RIF group had significantly less hospital stay than those in the ATO group. This interim analysis shows that oral RIF is as effective and safe as intravenous ATO for the treatment of pediatric APL, with the advantage of reducing hospital stay. Final trial analysis will reveal mature outcome data.


Subject(s)
Arsenic Trioxide/therapeutic use , Drugs, Chinese Herbal/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Adolescent , Arsenic Trioxide/administration & dosage , Arsenic Trioxide/adverse effects , Child , Child, Preschool , Disease-Free Survival , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/adverse effects , Humans , Infant , Length of Stay , Male , Treatment Outcome , Tretinoin/therapeutic use
12.
J Biol Inorg Chem ; 23(3): 313-329, 2018 05.
Article in English | MEDLINE | ID: mdl-29396610

ABSTRACT

Arsenic and its various forms have been in use in ancient Chinese medicine for more than 2000 years. Arsenicals have gained importance for having remedial effects for various diseases from syphilis to cancer thus highlighting its role as a therapeutic agent even though it has been labelled as a potential 'poison'. The ability of arsenic, specifically arsenic trioxide, to treat acute promyelocytic leukaemia has radically changed the perception of this poison and has been the main factor for the re-emergence of this candidate to Western medicine for the treatment of leukaemia and other solid tumours. This review highlights the glorious history of arsenic and its various forms with major emphasis on arsenic trioxide as a therapeutic agent. The mechanism of action, pathogenesis, pharmacokinetic profile, safety concerns, ongoing clinical trials and various new forms of arsenic trioxide are discussed. The review also outlines the therapeutic ability of this drug, discusses the latest developments and recent investigations and potential advancement of arsenic trioxide as nanoformulations that has made it emerge as a potential remedial agent.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenic Trioxide/therapeutic use , Neoplasms/drug therapy , Arsenic Trioxide/toxicity , Carcinogens/toxicity , Humans
13.
Leuk Res ; 65: 14-19, 2018 02.
Article in English | MEDLINE | ID: mdl-29232592

ABSTRACT

The aim of our study was to evaluate the impact of oral arsenic (the realgar-indigo naturalis formula, RIF) and all-trans retinoic acid (ATRA) on coagulopathy in acute promyelocytic leukemia (APL) compared with intravenous arsenic trioxide (ATO) and ATRA during induction. Mitoxantrone was added to all the patients at a dose of 1.4mg/m2 per day for 5-7 days. D-dimer levels, prothrombin time (PT), fibrinogen (Fbg) levels and the platelet count were comparably analyzed among 83 newly diagnosed APL patients treated with RIF (n=45) or with ATO (n=38). Since induction therapy with RIF and ATRA, the median levels of Fbg, PT and platelets were recovered to the normal range within 4days, 10days and 28days, respectively. The last day of platelet and plasma transfusion was day 12 (range: 0-24 days) and day 3 (range: 0-27 days), respectively. Among the 42 patients with a disseminated intravascular coagulation (DIC) score=4, the consumption of transfused platelets was less in the RIF group than that in the ATO group (P=0.037). In the 17 patients with a DIC score <4, prompt recovery of Fbg levels (P=0.028) was observed in the RIF group compared with that in the ATO group (P=0.401). RIF and ATO showed similar effects on the recovery of coagulopathy in APL patients. RIF had a potential beneficial effect in accelerating the recovery of thrombocytopenia and hypofibrinogenemia for subclinical DIC patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arsenic Trioxide/therapeutic use , Disseminated Intravascular Coagulation/drug therapy , Drugs, Chinese Herbal/therapeutic use , Leukemia, Promyelocytic, Acute/complications , Leukemia, Promyelocytic, Acute/drug therapy , Tretinoin/therapeutic use , Administration, Oral , Adolescent , Adult , Arsenic Trioxide/administration & dosage , Blood Transfusion , Disseminated Intravascular Coagulation/complications , Disseminated Intravascular Coagulation/physiopathology , Disseminated Intravascular Coagulation/therapy , Drugs, Chinese Herbal/administration & dosage , Female , Humans , Leukemia, Promyelocytic, Acute/blood , Male , Middle Aged , Mitoxantrone/therapeutic use , Platelet Transfusion , Retrospective Studies , Tretinoin/administration & dosage , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL