Next Article in Journal
Understanding the Binding Induced Folding of Intrinsically Disordered Proteins by Protein Engineering: Caveats and Pitfalls
Previous Article in Journal
A Novel 3D Bioprinter Using Direct-Volumetric Drop-On-Demand Technology for Fabricating Micro-Tissues and Drug-Delivery
Previous Article in Special Issue
Combination Drug Therapy for the Management of Alzheimer’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Overview of Dual-Acting Drug Methotrexate in Different Neurological Diseases, Autoimmune Pathologies and Cancers

by
Przemysław Koźmiński
*,
Paweł Krzysztof Halik
,
Raphael Chesori
and
Ewa Gniazdowska
Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(10), 3483; https://doi.org/10.3390/ijms21103483
Submission received: 4 April 2020 / Revised: 8 May 2020 / Accepted: 12 May 2020 / Published: 14 May 2020

Abstract

:
Methotrexate, a structural analogue of folic acid, is one of the most effective and extensively used drugs for treating many kinds of cancer or severe and resistant forms of autoimmune diseases. In this paper, we take an overview of the present state of knowledge with regards to complex mechanisms of methotrexate action and its applications as immunosuppressive drug or chemotherapeutic agent in oncological combination therapy. In addition, the issue of the potential benefits of methotrexate in the development of neurological disorders in Alzheimer’s disease or myasthenia gravis will be discussed.

1. Introduction

Methotrexate (MTX, amethopterin, Figure 1) is a synthetic organic compound, which belongs to an anti-folate therapeutic group and to class III of the Biopharmaceutical Classification System. It was created in the late 1940s as a less toxic derivative of aminopterin (Figure 1), then a folic acid (FA) antagonist used to treat children with acute leukaemia [1]. A few years later, it was noted that a low dose of aminopterin (1–2 mg/day) causes significant improvement in patients with rheumatoid arthritis (RA) [2] and in patients with psoriasis [3]. However, it was MTX that was introduced to clinical application in RA since the mid-1980s [4,5]. Currently, MTX is commonly applied in combination with other drugs for the treatment of many neoplasms (acute lymphoblastic leukaemia, acute myeloid leukaemia, meningeal leukaemia and lymphoma, osteosarcomas, non-Hodgkin’s lymphoma, also breast, bladder and number of other cancers) [6,7,8,9,10,11,12,13], severe and resistant forms of autoimmune diseases (rheumatoid arthritis, psoriasis, myasthenia gravis, Crohn’s disease, multiple sclerosis, polyarticular juvenile idiopathic arthritis) [4,5,6,7,14,15,16,17,18,19], or even an ectopic pregnancy [20].
Particularly, the introduction of low dose methotrexate (LDMTX) therapy of RA and psoriasis with dose of 7.5–25 mg/week versus high dose methotrexate (HDMTX) therapy of 1–5 g/week in cancer therapy became great breakthrough [15]. This approach was found to be relatively safe (especially in case of serious interactions with other drugs) and significantly reduced the occurrence of relevant adverse effects [6,7], what highly improved patient tolerance and therapy compliance. Since then, perception of MTX in the clinical environment has changed; moreover, this drug became the gold standard for the treatment of RA [21,22], demonstrating greater efficacy and safety than other synthetic disease-modifying anti-rheumatoid drugs (DMARDs), while biological drugs became only a complement to MTX application. The clinical success of MTX has prompted a further search for new multi-functional dihydrofolate reductase (DHFR) antagonists [23,24,25]. Over the past two decades, many natural and synthetic DHFR antagonists have been discovered and have already been registered mainly for oncological indications; however, MTX is still widely used in the treatment of various diseases and has not been allowed to become a thing of the past.
This review will present MTX in terms of its broad clinical use, application in the therapy of autoimmune diseases, including central nervous system disorders like myasthenia gravis (MG) or Alzheimer’s disease (AD) and application in oncological combination therapy with other drugs.

2. Methotrexate—Mechanisms of Drug Action

MTX is an anti-metabolite (anti-vitamin) of folic acid (FA, vitamin B9), which acts as anticancer agent and immunosuppressant [26,27]. MTX indirectly inhibits cell division through the blockage of folate-related enzymes, mainly DHFR, that catalyses the conversion of dihydrofolate to tetrahydrofolate (THF). THF serves as a significant coenzyme in several transmethylation reactions in pyrimidine and purine nucleotide synthesis pathways, essential in synthesis, repair or replication of DNA strands [28,29]. Actually, the methyl-THF acts as proximal methyl donor in numerous methylation reactions of DNAs, RNAs, proteins, phospholipids and amino acids syntheses. Inhibition of intracellular THF production by MTX results in disruption of cell proliferation and its metabolic imbalance.
MTX crosses the biological barriers very poorly, being highly ionized and generally hydrophilic. Bioavailability and biodistribution of the drug are determined by an active transport system [30,31]. Intestinal tissue adsorption of MTX occurs by the proton-coupled folate transporters (PCFTs), which are a solute carrier transporter, while a cellular drug penetration is followed mainly by the reduced folate carrier 1 (RFC1), an APT-binding cassette transporter. To a small extent, MTX also uses receptor-mediated endocytosis via folate receptors (FRs), the glycosyl-phosphatidyl-inositol (GPI)-anchored membrane proteins that may internalize bound folates and folate conjugates [32,33]. Intracellularly, MTX is metabolized by folylpolyglutamyl synthase (FPGS) to a polyglutamate derivatives (MTXGlu), that show significantly increased cell residence time and bioactivity in comparison to initial MTX form (Figure 2) [34,35,36]. This is a key pharmacokinetic step that determines the attributed effect of this drug, defining MTX as a representative type 1 prodrug, that undergoes bioactivation inside the cell [37]. Polyglutamated MTX is a superior anti-folate agent than MTX, capable of highly potent DHFR inhibition. Moreover, it also induces inhibition of other enzymes like thymidylate synthase (TYMS) [38], 5-aminoimidazole-4-carboxamide ribonucleotide transformylase (AICART) [39,40] and amido-phosphoribosyltransferase [41,42] participating in de novo biosynthesis of purine and pyrimidine nucleotides. Consequently, it is MTXGlu that deprives a cell of precursors for the synthesis of DNA and RNA necessary for cell proliferation (Figure 2), leading to DNA synthesis disturbances and subsequent cell apoptosis [43]. It should not be surprising that MTX activity is most visible in actively dividing cells, mainly in the S phase of the cell cycle, and in fact, that highly proliferating cancer cells are the most susceptible to the cytotoxic effect of this drug, indicating that antagonism of folate is related with the anti-tumour activity of MTX.
At the same time, the continuous action of MTX polyglutamates in cellular biochemistry results in intracellular accumulation of 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) by AICART inhibition. AICAR has an ability to diminish activity of adenosine deaminase (ADA) and adenosine monophosphate (AMP) deaminase (AMPDA) as well (Figure 2) [39]. An MTX-mediated excess of AICAR promotes the AMP and adenosine increase and subsequent release of these adenine derivatives outside the cell. Extracellularly, AMP follows easily dephosphorylation to adenosine via ecto-5′-nucleotidase, also known as the CD73 enzyme [44]. Adenosine, a significant signalling agent that modulates diverse physiological functions, acts as major mediator of anti-inflammatory action associated with MTX. In the local extracellular space, adenosine might interact with its specific receptors (A1, A2A, A2B or A3) present on the surface of the origin tissue, but also on immune system cells (Figure 2). Adenosine receptors belong to the seven-transmembrane receptor family, mediating signals into the cell by coupled specific G proteins depending on the receptor subtype. In RA patients, the occurrence of adenosine-specific receptors on immune and synovial cells is elevated, especially A2A and A3 that highly mediate adenosine regulation of immune response and inflammation [45,46,47,48]. The adenosine action on A2A and A2B stimulates intracellular production of cyclic adenosine monophosphate (cAMP), a significant cellular second messenger, while stimulation of A3 induces phospholipases C and D or inhibits adenylate cyclase producing cAMP. As a result, adenosine in activated neutrophils and macrophages reduces their ability of adhesion on endothelium and phagocytosis as well as generation of superoxide anions and further reactive oxygen species [49,50]. Moreover, leukocytes decrease the production of tumour necrosis factor α (TNF-α) and interleukin (IL)-12—the mediators of inflammation—and promotes anti-inflammatory IL-4 and IL-10. Additionally, it is observed that an inhibition of IL-1 action and suppression of TNF-α excretion in T cells and macrophages inhibit the prostaglandins and leucotrienes syntheses [50,51,52,53,54,55,56]. At the same time, adenosine disrupts the cytokine-mediated inflammation in endothelial cells by decrease in production of IL-6 and IL-8 and hindering leucocytes’ adhesion [57].
Thus, an adenosine-mediated effect seems to be a key mechanism in MTX anti-inflammatory action. Nevertheless, the MTXGlu also has some other impact on immune chemotaxis and reduction of the occurrence of inflammation mediators. Sustained decrease of THF-mediated methylation downregulates an accumulation of spermine and spermidine polyamines in the extracellular fluids and lymphocytes of patients with RA [58,59]. These polyamines are essential cellular growth factors among others in lymphocytes. MTX action inhibits T cell proliferation and synthesis of immunoglobulins or rheumatoid factor in RA patients [60,61], followed by a local reduction in lymphocytes interferon (IFN)-γ and IL-2 production, decrease of inflammation, convergent to adenosine receptor-mediated effects.

3. Methotrexate—Applications in Medicine

All of the above indicates that MTX, as a drug, has a pleiotropic mechanism of action. Potent anti-metabolic and anti-proliferating action is related with the therapeutic application of MTX as an anticancer agent. The action leading to a local increase of the adenosine concentration at the site of inflammation and also the specific immunosuppressive effect of MTX play an important role in the treatment of severe inflammation and autoimmune diseases (arthritis, psoriasis, myasthenia gravis). Moreover, MTX may serve as vector leading the MTX-attached agents to specific molecular targets like RFC or, less likely FRs [32,33,62,63], to be overexpressed on various cancer cells. While FA plays the role of a Trojan horse for molecular selective delivery of the FA-attached agent to FR-expressing cancer cells, MTX is considered the cornerstone of anti-folate therapy in the treatment of several malignancies.

3.1. Methotrexate—Anti-Inflammatory and Immunosuppressive Drug

Since the 1950s, MTX still remains a great interest as an anti-inflammatory and immunosuppressive drug. Its effectiveness in low dose therapy is so irreplaceable that it has become a basic drug of the disease-modifying anti-rheumatic drug (DMARD) category. Nowadays, DMARDs, like MTX, sulfasalazine, azathioprine (AZT) together with biological drugs (e.g., tocilizumab, infliximab) are used to relieve pain and stop disease progression. MTX is extensively used in rheumatoid arthritis, juvenile idiopathic arthritis, psoriatic arthritis, and other inflammatory disorders like myasthenia gravis (MG), Crohn’s disease, multiple sclerosis, sarcoidosis [64,65,66,67,68,69,70,71,72].

3.1.1. Methotrexate-Based Therapy of Myasthenia Gravis

MG, meaning “serious muscle weakness”, is a rare autoimmune-neurological disorder more commonly occurring in females than in males. It is manifested by weakness and rapid fatigue of pathologically altered skeletal muscles under voluntary control of the somatic nervous system [73,74,75]. MG usually starts in the extraocular muscles, especially those responsible for lifting of the eyelid. Often, only one eye is affected for a long time. Weakness quickly applies to facial muscles, palate muscles and extremities. In severe disease form, respiratory muscle involvement leads to breathlessness and life-threating breathing disorders [76,77].
Most patients with MG have abnormally elevated levels of antibodies against the acetylcholine receptor, muscle tyrosine kinase or lipoprotein-related protein 4 [78,79,80]. That is why treatments of MG are based on acetylcholinesterase inhibitors pyridostigmine and ambenonium to strengthen cholinergic neurotransmission. They are often combined with immunosuppressive drugs that reduce the overactivity of the immune system in production of antibodies against postsynaptic elements of neuromuscular junction. These drugs include glucocorticosteroids (GCS), AZT, cyclophosphamide and MTX [81], where GCS are the first-choice drugs applied for MG. In case of the absence of a response to the treatment, AZT may be applied or MTX as an alternative in AZT intolerance [82,83,84,85].
The study described by Hartman et al. gave information that LDMTX applied in patients suffering from MG was quite effective, safe and well tolerated [86]. A similar point of view is presented by Gold et al., where patients with MG received an MTX dosage of 7.5 to 15 mg orally per week [82,87]. MTX seems to have a beneficial effect in slowing progression of MG without any serious side effects. Abdou et al. showed some improvement in 87% of patients who received LDMTX intramuscular weekly for up to 20 months [88]. In another 24-month, single-blinded trial, authors compared LDMTX together with prednisone versus AZT combined with prednisone. They obtained comparable efficacy and tolerability results in both cases. Based on experimental data, the authors suggest that MTX may be a useful drug for MG treatment [89]. Karaahmet et al. described a significant improvement in the muscle weakness and fatigue of patients with MG after treatment with LDMTX over a year. The authors believe that MTX is an effective drug in the treatment of MG. However, they are aware that detailed research is needed to determine conclusive role of MTX in relation to MG [17]. A randomized, double-blind, placebo-controlled trial of MTX for patients with MG was described by Pasnoor et al. [90]. In this study, patients were treated with LDMTX and prednisone or placebo prednisone tablets. The obtained results showed no benefit of MTX in MG over 12 months of combination treatment. Based on the literature, MTX is a third-line agent for the treatment of MG.

3.1.2. Methotrexate-Based Therapy of Rheumatoid Arthritis

RA is an immune-mediated chronic disorder of connective tissue system manifesting in pain, stiffness and swelling of the joints, mainly of the hands and feet, but inflammation affects often other joints. No treatment leads to joint damage and severe disability, as well as damage to many organs and even premature death [91,92]. The aetiology of this disease is still not fully understood but involves a complex interplay of environmental and genetic factors. RA is a chronic disease originated by a specific antigen and caused by overreactive memory T cells, which play a key role in the non-specific inflammatory process. Self-activation of the memory T cells causes constant amplification of the disease. These lymphocytes release cytokines as IFN-γ and IL-2, which further activate monocytes and macrophages releasing subsequent IL-1, TNF-α and growth factors [93,94,95,96,97,98].
There are three main groups of drugs used for the management of RA: non-steroidal anti-inflammatory drugs (NSAIDs), corticosteroids and disease-modifying anti-rheumatic drugs. The first two groups aim only to reduce symptoms; however, modern therapy regimens are set to achieve disease remission introducing DMARDs early in treatment. Initiating treatment with DMARDs as soon as possible after diagnosis produces significant clinical and functional benefit for the patient.
MTX is commonly used for the treatment of patients with RA and in several other forms of inflammatory arthritis and autoimmune disease. According to many placebo-controlled trials of MTX versus other approved DMARD’s, MTX was established as the standard of care and first-line therapy drug for RA therapy. Furst et al. [99] confirmed that MTX with a dose of 12.5–20 mg/week had a significantly greater effect than the placebo in RA patients. A similar observation was described by Weinblatt et al. [100]. To increase the therapeutic effect, MTX is also used combined with other drugs like sulfasalazine, hydroxychloroquine and especially biological DMARDs. Based on literature, a combination of MTX with anti-TNF therapy was considerably better than monotherapy with MTX [101,102].

3.1.3. Methotrexate-Based Therapy of Alzheimer’s Disease

Alzheimer’s disease (AD), diagnosed by Alois Alzheimer in 1907, is a disorder of the central nervous system causing general dementia. Its causes and origins have not been fully understood. Alzheimer’s disease is most common in the elderly who are over 65 years old, and causes a decrease in in mental performance not directly related with age. The disease process causes damage to the cerebral cortex, which in turn leads to trouble with speech, memory or thinking. AD is a condition that develops very slowly and causes problems in everyday life over time. Ultimately, brain atrophy occurs, which is evident during computed tomography or magnetic resonance imaging.
Over the past years, a new hypothesis has emerged in which scientists point to inflammation as the primary cause of the AD. This hypothesis came from an observation that people with RA after use of NSAIDs had an unexpectedly low prevalence of dementia [103]. It is also notice worth that, according to the results published by researchers from the University of Oxford, UK, MTX can reduce risk for dementia, e.g., AD among patients with rheumatoid arthritis. They observed a significant reduction in risk for dementia among patients treated with MTX compared to non-treated patients. The therapeutic effect of MTX probably is caused by a lower dose than that for RA treatment, which is sufficient enough to cross the blood–brain barrier (BBB) to have an effect in AD protection [104]. According to the other research papers, MTX is associated with a lower risk for AD. Based on this knowledge, therapy with MTX shows promise application as a potential treatment for AD [105,106].

3.1.4. Methotrexate-Based Therapy of Other Diseases

Dermatology is another field where MTX is widely and successfully used [6,107,108,109,110]. As recommended, the drug can be orally administered or parenterally by injection and the first effects of MTX treatment usually appear after 2–6 weeks. In accordance with the literature, MTX with beneficial effects has been used in the treatment of other skin diseases, e.g., mycosis fungoides and lichen planus [111,112,113]. MTX has been also used successfully in the treatment of sarcoidosis as an alternative to corticosteroids [68,114,115]. Based on clinical trials, MTX is recommended as a first-line drug for patients with contraindication to corticosteroids because of toxicity or lack of efficiency [116].
Based on American College of Gastroenterology guidelines, MTX is effective and recommended for patients with steroid-dependent Crohn’s disease and for maintenance of remission. The published controlled trials comparing MTX with a placebo show that LDMTX intramuscularly combined with prednisone for 16 weeks or LDMTX monotherapy over 40 weeks, caused greater remission of the disease than the placebo [117,118]. Based on this knowledge, MTX may have a valuable role in selected patients with steroid-refractory or -dependent Crohn’s disease, even if MTX is not indicated by the US Food and Drug Administration (FDA).
Multiple sclerosis (MS), a potentially disabling disease of the brain and spinal cord, was first described in the 19th century by Jean-Martin Charcot [119]. Most often it concerns young people, with the peak incidence between 20 and 40 years of age, and a slight prevalence of cases in women than men.
MS is a disease where the immune system attacks neurons, oligodendrocytes, and brain immune cells in which the myelin sheath around the nerve cell appendages is damaged, thus causes inability to properly transmit impulses along the nerve pathways in the brain and spinal cord. The term "disseminated" reflects the spread of the pathological process to different places in the nervous system, as well as the interval of change over time. The disease usually has a multi-phase course, with periods of exacerbation and remission [120]
MTX is an off-label drug for multiple sclerosis treatment however, research has shown that it can reduce relapse rates and slow progression in people with MS, even if the mechanism of action of MTX is still a puzzle. A few trials of MTX have been conducted in multiple sclerosis patients. Goodkin et al. described that LDMTX orally has a slight benefit for MS patients compared to the placebo [19]. LDMTX has been also studied in comparison to IFN β-1α or with a combination of both, demonstrating that the treatment with MTX alone was effective, however combination therapy gives superior results [121,122]. A similar point of view is presented by Stark et al., where the MTX is described as a safe therapeutic option in advanced MS [123].
Information concerning MTX application as non-oncology drug is presented in Table 1.

3.2. Methotrexate—the Anti-Tumour Agent

Anti-folates are the first class of anti-metabolites introduced to the clinic approximately 60 years ago. Folate antagonists were among the first antineoplastic agents to be developed [63]. In 1948, the 4-amino derivative of FA (aminopterin) (Figure 1) was used to induce remission in childhood acute lymphoblastic leukaemia (ALL), and the related agent MTX is still an important component of modern cancer treatment [62,124,125]. MTX is FDA approved, alone or with other drugs, for ALL treatment both when it has spread to the central nervous system (CNS) or to prevent it from spreading there, as well as to treat people with other types of haematologic malignancies, e.g., advanced non-Hodgkin lymphoma (NHL) or advanced mycosis fungoides (MF). MTX is approved also to be used in the treatment of many other types of cancer like brain tumours, breast cancer, hepatoma, lung cancer, lymphomas, certain types of head, neck and esophagogastric carcinomas, gastric cancer, osteosarcoma that has not spread to other parts of the body or following primary tumour surgery, prostate and bladder cancers or gestational trophoblastic neoplasia [62,126].
However, MTX used in high doses in cancer therapy can also cause very serious, life-threatening side effects highly dependent on the treatment duration, age and condition of the patient. The minimal effective concentration and minimal toxic concentration of MTX is considerably small and the drug has a very narrow therapeutic range [127]. The risk of side effects is greater in patients regularly taking other medicines, especially NSAIDs like aspirin, ibuprofen or naproxen. HDMTX applied in oncological treatment, administrated for a long time, can cause a decrease in the number of blood cells and decrease of the activity of the immune system. HDMTX may cause respiratory failure and liver disease complications (especially in the elderly, obese or diabetic), serious skin reactions, lining damage of mouth, stomach or intestines and even foetal harm or death. Generally, during oncological therapy with HDMTX, the adverse reaction effects happen in more than 1 to 10 people (>10%). Therefore, MTX treatment requires close monitoring of the patient’s health and, if necessary, dose changes, FA or leucovorin (LV, folinic acid) rescue administration or even withdrawal from treatment plan altogether. Nevertheless, HDMTX with LV rescue has been most commonly used in a therapeutic strategy in oncology for almost three decades [126,127].
Concise information concerning MTX application (both alone and as a component of combination regimens) as an anti-tumour agent is presented below and in Table 2.

3.2.1. Methotrexate-Based Therapy of Haematologic Malignancies

Haematological malignancies (also called as liquid tumours) are a heterogeneous group of blood cancer (neoplastic diseases of the haematopoietic and lymphoid tissues), with clinical behaviour corresponding to leukaemia, lymphoma or myeloma (the subtypes of haematological malignancy according to World Health Organisation (WHO) classification) [124,125]. Leukaemia is defined as a progressive, malignant disease of the blood-forming organs, characterized by distorted proliferation and development of leukocytes and their precursors in the blood and bone marrow. Four subtypes of this malignance can be distinguished: ALL, acute myelogenous leukaemia (AML), chronic lymphocytic leukaemia (CLL) and chronic myelogenous leukaemia (CML). Lymphoma, defined as any malignancy of the lymphoid tissue, is distinguished into two main types: Hodgkin lymphoma and NHL. Both of these are of B-cell origin [239], while almost 90% of lymphomas are extranodal NHL and over 90% of lymphomas are of B-cell origin [124,178].
The most common form of T-cell lymphomas is cutaneous neoplasm MF, also known as Alibert-Bazin syndrome or granuloma fungoides. Multiple myeloma is a cancer of the plasma cells. The malignancy of haematologic neoplasms ranges from relatively indolent to highly aggressive.
MTX as a cytostatic drug and is widely used to treat many cancers of haematological origin alone or in combination regimens with other anticancer agents (Table 2). The first applications of MTX were used in the 1950s/1960s for childhood ALL treatment [128,167,168,169,170]. Patients were given repeated infusions of HDMTX followed by LV rescue administration. Despite numerous studies conducted in many patients, intravenous (i.v.) HDMTX has not been proved conclusively to be more effective than less toxic and less costly methods based on LDMTX [128]. The promising and effective result of HDMTX application has been described by Sakura et al. (a randomized phase III trial comparing HDMTX therapy with intermediate-dose (IDMTX) therapy) [166]. Adult patients with Philadelphia chromosome negative ALL were randomly assigned to receive therapy containing HDMTX (3 g/m2) or IDMTX (0.5 g/m2). The estimated 5-year disease-free survival rate was 58% and 32% in patients treated with HDMTX and IDMTX, respectively.
A deep analysis of efficacy of HDMTX application in the treatment of children with ALL is presented in the paper of Gong et al. [177]. Children with ALL were divided into smaller subgroups according to the subtype of the disease (B- or T-lineage ALL) and disease course. At different time periods after HDMTX infusion, the MTX plasma concentrations and adverse reactions were tested and compared. The obtained results showed that MTX plasma concentration in the B-lineage group was significantly higher than for T-lineage group and the incidence of adverse reactions in the high-risk group was significantly higher than for the moderate- and low-risk groups. HDMTX caused more adverse reactions in B-lineage ALL children compared to T-lineage ones. During clinical application of HDMTX in the treatment of ALL, particular attention was paid to changes in the vital signs of patients and, if necessary, calcium formyltetrahydrofolate or citrovorum factor rescue was administrated.
MTX application in lymphomas are reported in numerous papers. In this work, we selected the most comprehensive articles on MTX-based lymphoma therapy (Table 2) [124,129,171,172,173,174,175,176,179,182,240,241]. Primary central nervous system lymphoma (PCNSL) is a malignant disease of the lymphatic system that accounts for about 2% to 5% of all primary intracranial tumours in immunocompetent patients. It is an aggressive NHL of B-cell origin and can be located in the brain, meninges, eyes, spinal cord, cerebrospinal fluid and intraocular structures [178,182]. Studies of the use of HDMTX as a single agent followed by LV rescue in patients with widespread NHL have demonstrated the effectiveness of this treatment [171,172,173,174,175,176]. Zhu et al. studied the response and adverse effects of intravenous HDMTX administration as a function of age at the time of PCNSL diagnosis [176]. The obtained results showed no significant age-dependent differences in the median progression-free survival (PFS) or overall survival (OS), but higher risk of treatment-related complications in elderly PCNSL patients because of the increased prevalence of comorbidities. MTX in low doses as an anticancer agent is rarely used alone [129,130]. It is usually used in combination regimen with other cytostatics. Wen et al. tested clinical efficacy and toxicity of metronomic administration of LDMTX and thalidomide in relapsed and refractory NHL [136,137]. Generally, the therapy was well tolerated and most patients achieved an objective response (OR).
The most common MTX-based chemotherapy in lymphoma treatment is combination regimen containing HDMTX and other anti-neoplasm preparations [131,132,179,180,181,183,184,185,186,187]. Applications of combination regimen containing HDMTX and cytarabine (Ara-C) for systemic NHL treatment have been described by Bergner et al. [179] and Bokstein et al. [180]. The obtained results indicated a benefit in patients treated with combined HDMTX+Ara-C chemotherapy compared to patients treated with HDMTX alone; however, HDMTX+Ara-C chemotherapy was also less tolerated. Nevertheless, the combination of HDMTX and a high-dose Ara-C regimen is today the backbone of the first line therapy of PCNSL [178]. MTX and temozolomide combination for the elderly suffering from PCNSL treatment was tested by Omuro et al. [181]. Among the patients, a complete response was observed in 55%, and the disease progressed in the other 45%.
Other HDMTX-based examples of combination regimens used in patients with lymphoma treatment are bleomycin, Adriamycin, Cytoxan and vincristine [172]; rituximab, CTX, doxorubicin, vincristine, prednisolone (R-CHOP) and intrathecal MTX [183]; rituximab, MTX, procarbazine and vincristine (R-MPV) [184]; MTX and rituximab [185,186,187]; rituximab, MTX, Ara-C, dexamethasone (R-MAD) [182]. Generally, outcomes of PCNSL treatment with HDMTX-based regimens remain poor, presumably due to the low ability of MTX to penetrate CNS. In addition, MTX has limited use in cancer chemotherapy because of its dose-dependent adverse effects, such as poor bioavailability, toxicity, low specificity, and drug resistance. The use of combination regimens in most therapies led to an increase in complete response rate and a significant prolongation of PFS compared with the results obtained in HDMTX monotherapy.
LDMTX alone [131,132,133,134] or in association with other chemotherapeutic agents is recommended for the treatment of primary cutaneous CD30-positive lymphoproliferative disease, as well as alone [131,132,133,134] or in combination with interferons [131,144,145,146,147] recommended for treatment of refractory MF neoplasm. The lateral combination regimen resulted in a very high clinical efficiency and durable response without serious adverse reactions [131,144,145]. In the 1980s, MTX was used to MF treatment in combination chemotherapy containing bleomycin and doxorubicin or along with topical nitrogen mustard [147]. The lateral combination turned out to be quite effective for the treatment of MF advanced stages. Similarly effective for advanced MF treatment was a regimen containing MTX and fluorouracil (5-FU) [189]. MTX administration followed by 5-FU and LV rescue was well tolerated and did not cause any serious side effects. Some years ago, Kuo et al. reported the positive results of the treatment of erythrodermic using LDMTX and UV-B irradiation [148]. The use of MTX in another therapeutic application was presented by Raychaudhury [149]. LDMTX along with radiotherapy including total skin electron beam therapy is recommended in India for intermediate-risk patients with MF.

3.2.2. Central Nervous System—Methotrexate Brain Cancer Therapy and Brain Drug Delivery

Brain tumours can be divided into two main types: malignant or benign tumours. The limited success of chemotherapy against brain tumours is caused mainly by low CNS drug penetration through the BBB [242]. MTX brain penetration is poor and due to its application at LDMTX is insufficient to achieve therapeutic concentration in the brain [190,191,243,244,245]. In order to attain the desirable MTX concentration in brain cancer, administration of HDMTX is required and current efforts in the field of MTX brain tumour therapy focuses mainly on the design of novel, non-invasive, safe and effective MTX delivery systems for cerebral treatment [199,246,247,248]. An important role in this area play a nanoparticulate (NP) delivery systems investigated with the aim of targeting therapeutics to the brain [199]. Although MTX has been used in medicine for decades, this type of delivery systems began to appear only for several years simultaneously with rapid development in the field of nanomedicine. Many novel approaches of MTX delivery to the CNS are presented below [63,200,201,202,203,204,205,206,207,208,209,210].
Based on the knowledge that some endogenous amino acids are able to cross the BBB, it can be considered that these amino acids can be used as carriers in molecules transported to the brain. Singh et al. designed reversible conjugates MTX-(glutamine)2 and -(lysine)2 to enhance the brain availability of MTX [190,191]. The biodistribution studies showed much higher accumulation in the brain of both compared to that of free MTX, indicating effective transport of novel conjugates across the BBB and more effective MTX brain penetration.
Corem-Salkmon, in the frame of novel approach to deliver drugs directly into brain tumours, presented the application of biodegradable magnetic nanoparticles (NPs) with MTX, conjugated with the human serum albumin (HSA) [192,193]. Studies on formulations of MTX-HSA have been described also by Wosikowski et al. [194] and Burger et al. [195]. The study results showed that this approach can alter MTX pharmacokinetic, enhance tumour targeting, reduce toxicity, overcome drug-resistance mechanisms and extend the clearance time of the conjugate. Kohler et al. designed, synthesized and studied magnetic iron oxide nanoparticles conjugated with MTX [196]. This preparation can be used both as a diagnostic agent in magnetic resonance imaging and as a carrier in controlled drug delivery. Gao and Jiang studied the transport ability of MTX-loaded polyacrylate nanoparticles [197]. MTX NPs, especially of diameter below 100 nm, had the ability to cross the BBB. Application of such conjugates significantly improved MTX level in both brain tissues and cerebrospinal fluid (CSF). Trapani et al. described the synthesis and physicochemical properties of formulation of MTX with chitosan NPs [198]. These nanocarriers seem to act as MTX reservoirs, allowing slow drug release in the tumour area, once the BBB is crossed [192,198]. Another MTX-loaded NP described in the literature is based on the biodegradable polymers: polyesters of (poly(lactic acid) (PLA) and poly(d,l-lactide-co-glycolide) (PLGA) [199,249]. Both these polymers are preferred as components for formulations indicated for brain drug delivery as the non-toxic products of their autocatalytic hydrolysis are degraded to CO2 and H2O. Studies of physicochemical and biological properties of MTX-polymeric NPs showed that these MTX-loaded NPs improved MTX delivery to the target, what increased therapeutic effect and reduced serious adverse effects on normal tissues such as kidney and liver. An MTX-based polymeric delivery system can be also prepared from proteins (gelatine, albumin) [199,200,201,202,203,204,205,206]. Narayani and Rao presented biodegradable hydrophilic MTX-gelatine microspheres [201,202]. The rate of MTX release from microspheres decreased with increase in the particle size of the microspheres and was faster in gastric fluid than that in intestinal fluid. Another MTX formulation in the form of gelatine microspheres was studied by Pica et al. [203,204]. These preparations, intended for slow drug release directly within the tumour, have been designed to minimize the systemic toxic effects of MTX and to overcome tumour resistance. However, contrary to expectations, the molecules of free MTX were released more efficiently from microspheres containing less tethered drugs. Hydrophilic biodegradable gelatine microspheres with covalently attached MTX provided prolonged drug release and increased anti-tumour activity. Silica-gelatine hybrid aerogels [205] are excellent candidates for controlled MTX delivery. Application of these aerosols can provide the same therapeutic effect inside the tumour site as free MTX, but without any adverse effects.
A promising platform applied in nanomedicine for the design of new drug delivery systems is dendrimers [206,207,208,209]. Application of MTX formulations built on generation 5 (G5) acetylated poly(amidoamine) (PAMAM) dendrimers have been tested and described by several research groups [206,207,208,209]. Wu et al. described synthesis as well physicochemical and biological characterization of multi-component conjugate C225-G5-MTX containing monoclonal antibody cetuximab (also called as C225), covalently linked to G5 PAMAM dendrimer, and previously coupled with a cytotoxic drug MTX [208]. This formulation is an effective way to deliver the drug to the target tissue, and can also increase the therapeutic index of MTX. The formulation G5-MTX containing MTX molecules tethered in the PAMAM G5 dendrimer makes MTX action about 10 times stronger and much less toxic than in the case of drug alone application [209]. Conjugates of MTX molecules and the fifth-generation PAMAM dendrimer, G5(MTX)n, where n = 0÷12, were synthesized and studied by Wong et al. [62]. Based on the obtained results, the authors concluded that these conjugates bind to FRs three to four orders of magnitude stronger than free MTX and that the drug molecules tethered in dendrimer can still inhibit a human DHFR enzyme as potently as free MTX does; however, this potency strongly depends on the length of applied in the syntheses linkers between dendrimer and MTX molecules. The studies have also showed the effectiveness of G5(MTX)n conjugates application in a tumour-targeting nanodelivery strategy. Another dendrimer-based MTX conjugate has been described by Dhanikula and Hildgen [210]. In order to evaluate the potential application of these drug delivery systems, the authors studied the efficiency of MTX encapsulation and release from polyester-co-polyether (PEPE) dendrimers. Riebeseel et al. designed and studied hybrid conjugates (MTX-PEG) of MTX and poly(ethylene glycol)s (PEGs) [211]. PEGs are potential drug carriers for improving the therapeutic index and delivery system of anticancer agents. All of the MTX-PEG conjugates inhibited DHFR in a similar manner to free MTX, but they were slightly less active than free MTX. The level of cytotoxicity of these preparations decreased with increasing sizes of MTX-PEG polymer conjugates.
It is also worth mentioning that the MTX-EPR conjugate combines two significant anticancer strategies: combinatory therapy and targeted therapy [212]. The MTX-EPR hybrid molecule contains two anticancer drugs, MTX and epirubicin (EPR), and each of them is characterized with a different mechanism of action. Co-administration of these two drugs in combination therapy causes the appearance of a synergistic effect. In addition, the cytotoxic effect of such a hybrid molecule is lower than in the case when both chemotherapeutics are used in therapy, but they are not administrated simultaneously.
Concise information concerning MTX-based therapy of CNS are presented in Table 2.

3.2.3. Methotrexate-Based Therapy of Head and Neck

Head and neck squamous cell carcinoma (HNSCC), cancer that arises in the nasal cavity, sinuses, lips, mouth, salivary glands, throat or larynx, is the 10th most common tumour worldwide. These sarcomas are rare cancers, and due to their variable clinical behaviour, location in close proximity to important vital structures and diversity of histologic subtypes, these neoplasms are difficult to diagnosis and effective treatment. Nowadays, in the era of personalized medicine, head and neck carcinomas belong to a critical oncological topic. Conventional HNSCC chemotherapy consists of drugs administration in cycles near or at the maximum tolerated dose, followed by an appropriate drug-free period allowing the patient to recover from acute drug toxicities. MTX, particularly in a high dose with LV rescue, is one of the main cytostatics used in head and neck carcinoma therapy (Table 2). It is used alone or in a combination regimen with other drugs; however, head and neck cancer therapy also requires radiation therapy and/or surgical tumour removal. MTX in different treatment regimens has been used in HNSCC therapy for several decades [138,139,188,250,251,252,253,254,255,256,257]. The use of MTX alone is described in numerous works [139,211,250,253,254,258,259,260,261,262]. MTX in different combination regimens is considered as a first-line drug indicated for palliative care in the case of head and neck carcinomas [261,262]. However, monitoring the patient condition is particularly important here, as patients are often elderly people with a multitude of comorbidities.
MTX alone or in combination with other drugs is used in metronomic chemotherapy in metastatic HNSCC patients [140,141,263,264]. Pai et al. described the application of LDMTX and celecoxib in locally advanced (stage III and IV) oral cancers [141]. The treatment was carried out twice before the surgery and during the perioperative period. This turned out to be more effective compared to cisplatin (PDD, cis-diammindichloridoplatin) reference chemotherapy. The authors documented in their work the advisability of using MTX in the neoadjuvant and adjuvant setting in advanced HNSCC. Mateen et al. described metronomic chemotherapy procedure for head and neck cancer therapy of oral LDMTX and capecitabine for at least a period of six months after chemoradiotherapy [143]. Based on the recorded results, the authors concluded that this regimen is an effective treatment option for recurrent HNSCC.
MTX is used from the past to now as a component in many combination regimens. Bertino et al. [203] tested the efficiency of the combination of MTX and bleomycin; these attempts have been discontinued due to high side effect toxicity. Next, the authors tested the effects of simultaneous administration of MTX and 5-FU. Application of HDMTX with bacilli Calmette Guerin vaccine described by Buechler et al. [188] provided only a temporary response. The combination regimen of MTX and LV with cyclophosphamide and cytosine arabinoside (MLCC) is presented in work of DeConti et al. [255]. Application of this regimen compared to MTX with LV resulted in more hematologic toxicity. The aim of the study conducted by Hong et al. was to compare MTX therapy versus PDD in patients with recurrent HNSCC [256]. The obtained results showed comparable effects of both cytostatics, however MTX proved to be better tolerated. In addition, comparison of the efficiency of MTX and a combination regimen containing a high-dose of PDD, Oncovin (vincristine) and bleomycin (COB) is discussed in the paper of Drelichman et al. [257]. Therapeutic effects of both sets turned out to be comparable, however nausea and vomiting were the more common side effects of COB, while hematologic toxicity was more frequent and more severe in the case of MTX application. Another therapeutic kit containing MTX, LV, PDD and 5-FU was tested by Chang et al. [139]. This regimen showed total safety profile and improved survival in patients with metastatic/recurrent HNSCC. Kushwaha et al. compared therapeutic effect observed in recurrent HNSCC treatment of three different regimens: gefitinib, MTX or MTX combined with 5-FU [140]. The authors observed comparable median survival rate in all cases, however a different toxicity profile. Adverse toxic effects oral mucositis, nausea, vomiting and haematological toxicity were recorded in the case of regimens of MTX and MTX with 5-FU while cutaneous rash, diarrhoea and oral mucositis were observed in the case of gefitinib. MTX or afatinib application in the treatment of HNSCC was studied and evaluated by Guo et al. [135]. Afatinib proved to give a significant improvement in patient PFS, slightly better than MTX. Very optimistic test results were obtained in the treatment of advanced/recurrent HNSCC patients using metronomic chemotherapy of MTX and celecoxib combination [142]. Celecoxib enhanced the therapeutic efficiency of MTX, treatment was well tolerated and provided good pain control and improves quality of life.

3.2.4. Methotrexate-Based Therapy of Breast Cancer

Breast cancer is the most frequently diagnosed life-threatening cancers in women and the main reason of death among women with cancer. MTX is used (alone or in combination) to treat both early-stage (as well after surgery and other treatments) and advanced-stage breast cancer. Some selected application of MTX in breast cancer therapy are presented below [196,213,214,215,216,217,218,219,220,221,244,265,266,267], Table 2. Martin et al. described numerous combination regimens applied in metastatic breast cancer, such as CTX, MTX, 5-FU (CMF), 5-FU, Adriamycin, CTX (FAC) and MTX in combination with tegafur and uracil followed by LV rescue (MUL) [213,214]. The authors demonstrated the superiority of MUL regimen compared to other ones. This regimen stopped disease progression in nearly 75% of patients. MUL chemotherapy turned out to be active and well tolerated and the toxicity was mainly gastrointestinal. Application of the regimen containing MTX and CTX in patients with metastatic breast cancer is described by Colleoni et al. [218]. Continuous administration of both at very low doses over a long period of time without treatment interruptions (typical for anti-angiogenic therapy) was well tolerated. The same research group presented the benefit of application of combination regimen CMF followed by tamoxifen or tamoxifen alone, as adjuvant chemotherapy in women with lymph node-negative breast cancer [266]. Based on experimental data, the authors showed that the classical CMF regimen is safe and plays a key role in the response to therapy in early breast cancer [215,216]. Cocconi et al. compared CMF regimen with the modified CMF method they proposed. The new regimen, CMFEV, contained CMF components and epirubicin or vincristine, rotationally [216]. The study demonstrated superiority of the CMFEV regimen over CMF. CMFEV showed greater anticancer activity in premenopausal patients and more effective in terms of long-term outcomes. Leone et al. described applications in breast cancer therapy for the regimens CMF and FAC [217]. The aim of the study was to describe the long-term results of these regimens application in neoadjuvant chemotherapy in stage III breast cancer patients. Both regimens were well tolerated and showed similar objective response (OR), long-term toxicity, disease-free survival (DFS), and overall survival (OS) rate at 16 years. Wu, C. E. et al. described a CMF regimen application in adjuvant chemotherapy of triple-negative breast cancer (TNBC), characterized with negative tests for oestrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2) [267]. This kind of therapy turned out to be effective in reducing recurrence amount and prolong DFS in patients with node negative TNBC, particularly in the case of tumours more than 2 cm or tumours after partial mastectomy. Fukuda et al. described a retrospective analysis of the efficacy of drugs mitomycin C and MTX in HER2-negative patients with metastatic breast cancer [219]. They described also the combination chemotherapy of mitomycin C and MTX (MM) in patients previously treated with aggressive anthracyclines, taxanes, capecitabine and vinorelin (ATCV) regimens. Under these conditions, MM chemotherapy proved to be effective and tolerated, however, only in patients who kept good performance status and bone marrow function even after multiple chemotherapy regimens. The combined effects of MTX and 5-aminoimidazole-4-carboxamide riboside (AICAR) on tumour cell proliferation were the object of research conducted by Cheng et al. [220]. In the frame of these researches, the influence of MTX on AICAR anticancer action and pharmacokinetics in human breast cancer as well as concentration of active AICAR metabolites in plasma and tumours were studied. The authors recorded a synergistic effect of MTX and AICAR combination on the inhibition of tumour cell proliferation—the combination therapy displayed more potent cytotoxicity against tumour cells than either drug alone. The synergistic effect of anti-tumour action was observed also in TNBC treatment reported by Wu, C. W. et al. [221]. They proposed the simultaneous use of MTX and a low dose of vitamin C. Both drugs taken separately do not have the ability to inhibit the growth of TNBC cells, however, the combination of vitamin C and MTX had synergistic anti-proliferative and cytotoxic effects on TNBC cells.
Breast cancer usually spreads to the CNS inducing commonly leptomeningeal metastasis. Kapke et al. determined that systemic intravenously administration of HDMTX, improves quality of life and provides durable remissions for leptomeningeal metastasis of breast cancer [244]. Application of MTX in a form of iron oxide NPs for human breast cancer therapy is given in the paper of Kohler et al. [196]. The release of MTX inside the tumour cells occurred as a result of the drug cleaved from the NPs under intracellular low pH conditions in cancer cells. Kumaki et al. described the case of two breast cancer patients with a history of LDMTX treatment for RA, who had histological findings similar to those seen after neoadjuvant chemotherapy [268]. The authors showed that oral LDMTX induced histological results similar to those seen in HDMTX chemotherapy, such as metronomic chemotherapy.

3.2.5. Methotrexate-Based Therapy of Lung Cancer

Lung cancer is one of the most common types of cancer, and according to current knowledge, the occurrence of this tumour is closely related to smoking [269]. Worldwide, over 1 million people are diagnosed with lung cancer each year. There are two main categories of lung cancer: small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). NSCLC (any type of epithelial lung cancer) accounts for about 85% of all lung cancers. Lung cancers are primarily treated by surgical resection, although chemotherapy (among others with MTX) is also used both pre-operatively (neoadjuvant chemotherapy) and post-operatively (adjuvant chemotherapy). In general, NSCLC is relatively insensitive to chemotherapy, while SCLC has a faster growth rate, shorter remission times and previous metastases to various locations in the body (e.g., cervical, prostate, gastrointestinal tract). According to the literature data, MTX is relatively rarely used in lung cancer treatment, however the examples of its application in lung cancer therapy have been described in several papers (Table 2).
Vincent et al. described and compared the use of a single drug MTX and five different combination regimens in SCLC chemotherapy [151]. The regimens always contained the drugs MTX, CTX and lomustine and one of the following chemotherapeutics etoposide, Adriamycin or vincristine. The most effective regimen was a set containing all of the above-mentioned drugs followed by radiation therapy to the chest. The therapy was well tolerated by patients, and provided a high objective tumour response rate. Handle et al. presented SCLC therapy based on two regimens—HDMTX or LDMTX with LV rescue in combination with CTX, doxorubicin and vincristine cycles alternating with etoposide, vincristine and hexamethylmelamine cycles [152]. In both approaches, the recorded response rates, median survival and overall survival were comparable, but adverse effects were more common in the case of HDMTX application. Overall, the HDMTX-based approach was less effective and associated with a higher cost and toxicity. Therapy of NSCLC with HDMTX followed by citrovorum rescue, mechlorethamine and procarbazine was studied by Bhasim et al. [222]. Although HDMTX followed by citrovorum rescue was relatively well tolerated, the addition of these two medications failed to improve the tumour response rate and increased toxicity. Another combination regimen containing LDMTX or HDMTX and drugs vincristine (Oncovin), doxorubicin and CTX (MOAC) for SCLC treatment are presented in the work of Neijstrom et al. [153]. Although HDMTX was able to achieve potentially therapeutic concentrations of MTX in CSF, the results of the study were not conclusive. There were no statistically significant differences in response rate or survival between these two approaches and there was no basis to conclude that HDMTX could prevent brain metastasis or the appearance of other CNS diseases. Interactions of LDMTX and PDD in NSCLC treatment were studied by Preiss et al. [154] and results here were not conclusive as well. Contrary to expectations, the authors observed no significant change in the kinetics of PDD in plasma after co-administration of MTX. After combined drugs administration, no signs of nephrotoxicity were observed and the overall toxic effects were on a mild level. Malzyner et al. studied, evaluated and compared numerous combination regimens used in advanced NSCLC therapy [155]. The authors described two pairs of sets: the MVP set containing mitomycin C, vinblastin and PDD and the M-MVP set containing additional MTX, the MNP set containing mitomycin C, vinorelbine and PDD, and the M-MNP set containing additional, as before, MTX. Based on the obtained results, Malzyner et al. reported superiority of the M-MVP regimen compared to MVP and the M-MNP compared to M-MVP. The M-MNP regimen, characterized with high positive response rate, as well as mild and manageable toxicity in patients.
Lung metastasis is the most crucial event affecting the treatment of osteosarcoma and is strongly dependent on tumour angiogenesis. With this in mind, Tomoda et al. investigated the inhibitory effect of MTX on lung metastasis, and on angiogenesis associated with the early stage of tumour formation [270]. The study was performed using a rat osteosarcoma S-SLM cell line with high metastatic potential. The obtained results showed that LDMTX, through its anti-angiogenic activity, significantly suppressed lung metastasis. In their work, the authors also drew attention to the fact that, although HDMTX is commonly used for the treatment of osteosarcoma, the conventional MTX dose in this therapy is ineffective. In the experiments performed in the frame of this work, low MTX doses could not inhibit the growth of tumours and kill osteosarcoma cells; however, they had a potency to suppress the lung metastasis process. Based on these facts, the authors concluded that MTX mechanisms of anti-angiogenic action and cytotoxicity are different.
Chen et al. designed and tested a new formulation MTX-PMX-PCNPs in which two different drugs MTX and pemetrexed (PMX, novel anti-folate with indication for lung cancer and malignant pleural mesothelioma treatment) were bonded with PEGylated chitosan nanoparticles (PCNPs) [223]. Drug release profiles of MTX and PMX were investigated using LLC and A549 cell lines. The obtained experimental results showed a stronger overall effect of the MTX-PMX-PCNP conjugate on the decreasing cell proliferation than the combined effects of MTX and PMX used separately, indicat that MTX and PMX had synergistic anti-tumour activity. The IC50 value of the formulation was significantly lower than those of free drugs. The use of MTX and a potent tubulin-binding anti-tumoural drug pretubulysin, alone and in the form of a combined regimen, is described by Kern et al. [224] The authors demonstrated the benefits of using the drug regimen and indicated that it was a promising therapeutic approach for various types of cancer, among other in lung cancer therapy. Sekimura et al. presented a case of primary lung cancer with MTX-associated lymphoproliferative disorder [271] which occurred in a patient receiving oral MTX and prednisolone for RA for about 15 years. Lung cancer therapy in such cases requires discontinuation of MTX, accurate diagnosis and initiation of other kind of treatments. Yan et al. noted the influence of acetylsalicylic acid (ASA) on the therapeutic anticancer activity of MTX [225]. ASA is a non-selective cyclooxygenase inhibitor that takes part in the treatment of inflammatory conditions such as, for example, RA. The anticancer activity of MTX and ASA were tested both alone and in combination, using two cell lines of human NSCLC, CL1-0 and A549. Based on experimental results the authors demonstrated that ASA suppresses the therapeutic efficacy of MTX in human lung cancer treatment via preserving cancer cell proliferation and survival. They concluded that the efficacy of MTX anticancer therapy can be decreased by ASA and, due to that application, these two medications together should be avoided. The novel and effective way of MTX pulmonary delivery in case of lung cancer treatment has been designed by Abdelrady et al. [226]. The authors designed and tested the application of MTX-gelatine nanoparticles obtained in different MTX loading techniques. The new preparation was well tolerated and had low toxicity, but most importantly, a satisfactory therapeutic effect was achieved with four times lower doses of MTX than in the case of standard MTX-based lung cancer treatment.

3.2.6. Methotrexate-Based Therapy of Prostate and Bladder Cancers

Urothelial cancer (UC) is the fifth most common malignancy both in men and women (the male to female occurrence is 3:1). Approximately, more than 400,000 new cases are being recorded annually and approximately 150,000 die from this disease, worldwide. Bladder cancer is a common urologic cancer that has the highest recurrence rate of any malignancy. MTX has been used for bladder cancer treatment since the 1970s to the 1980s of the last century [10,156,157,158,159,160,161,162,163,227,228,272]. It was used both alone [10,156,158,228] and in combination regimens with such antineoplastic agents as PDD, vinblastine, Adriamycin, 5-FU, vincristine, bleomycin, mitomycin C or cyclophosphamide [10,159,160,161,162,272]. MTX used as a single agent is one of the most active drugs [161].
One of the first applications of MTX-based treatment of locally advanced or metastatic transitional cell carcinoma (TCC) of the bladder are presented in the works of Hall et al. [156] and Turner et al. [158]. Hall’s research showed objective evidence of tumour regression in 26% of patients and several other patients gained relief of disease symptoms, but in these there was no clear evidence of tumour regression. Occurring toxic side effects associated with MTX application caused treatment interruption for a time in most patients [156]. Turner et al. described studies in which MTX was used in patients with advanced bladder cancer that did not respond to previously applied radiotherapy or had recurred after radiotherapy, and patients with metastatic disease. Patients were treated with LDMTX i.v. with or without LV rescue [158]. The obtained results show high effectiveness of these therapies, while the intensity of the side-effects was related with LV rescue. Generally, the medical experiments described above showed that PDD therapy is of value in the management of advanced bladder cancer. Natale et al. reported results of patients suffering from transitional urothelial tract tumours treated with LDMTX i.v. with or without citrovorum factor rescue [10]. Based on the obtained results, authors concluded that MTX in the treatment of patients with advanced urinary bladder cancer is as active as PDD. In another therapeutic approach, patients with advanced primary bladder cancer and metastatic lesions were treated using combination regimen consisting of MTX, 5-FU, vincristine, bleomycin and mitomycin C [227]. Complete and partial responses were recorded in the case of 9% and 23% of patients, respectively. HDMTX-based treatment with LV rescue of patients with TCC of the bladder has been described by Hall et al. [228]. The authors concluded that transurethral resection followed by HDMTX application may offer an effective alternative to radiotherapy or cystectomy for many patients with invasive bladder cancer. Kaye et al. described the results of the patients with advanced bladder cancer using MTX alone or in combination with PDD [159]. The obtained results showed that patients with satisfactory renal function can receive low doses of both drugs simultaneously without risk of enhanced toxicity. MTX is used as a component of numerous anticancer combination regimens [160,161,162,163,164], while the most commonly used in the advanced bladder carcinoma are MTX-vinblastine-Adriamycin-cisplatin (MVAC), HDMTX-vinblastine-Adriamycin-cisplatin (HDMVAC) and cisplatin-MTX-vinblastine (CMV). Since 1990, the MVAC and HDMVAC have been considered as standard first-line therapies in this indication [162]. MVAC showed superior effectiveness than single agent PDD, despite increased toxicity [162,163,164,165].

3.2.7. Methotrexate-Based Therapy of Osteosarcoma

Osteosarcoma is the most common histological form of primary bone cancer that is characterized histologically by the production of osteoid by malignant cells [165,273]. This neoplasm is most prevalent in children and young adults (it accounts for about 3.4% of all childhood cancers and 56% of malignant bone tumours in children). It is characterized by local infiltration and early, distant, hematogenous metastasis [236]. HDMTX with LV used alone or in different regimens exhibit significant activity against relapsed and metastatic osteosarcoma. In the work of Jaffe et al., four combination regimens of HDMTX-LV with doxorubicin, PDD, ifosfamide or CTX utilized in osteosarcoma treatment are presented [229]. As a main adverse effect, the authors mention renal failure caused probably by MTX precipitation in the renal tubules. Other toxic effects were gastrointestinal, hematologic, and hepatic dysfunction. Xu, M. et al. analysed the clinical efficacy of the cisplatin-ifosfamide-doxorubicin (DIA) regimen in osteosarcoma treatment and compared this therapy with HDMTX [274]. DIA therapy turned out to be better tolerated. Bielack et al. described the research conducted by European and American Osteosarcoma Study Group. They tested the final effect of therapy in which the treatment process was extended by the additional stage including treatment with MAP regimen (containing MTX, doxorubicin and PDD) enriched with the PEGylated formulation of IFN-α-2b [230]. The obtained result was not conclusive, because the slightly more effective final result of the extended therapy was not statistically different from the effect of the therapy without this additional stage. The problem of appropriate dose indication for MTX as a component of a multidrug regimen for the osteosarcoma treatment was tested by Krailo et al. In the frame of a randomized clinical trial in postoperative chemotherapy in the treatment of childhood osteosarcoma, two regimens were tested and compared: Adriamycin and vincristine combined with HDMTX or a moderate MTX dose. Both therapies showed similar effectiveness [231], which allows the conclusion that a moderate dose of MTX is sufficient in postoperative multidrug adjuvant chemotherapy of osteosarcoma. Mayers et al. investigated the therapeutic effect resulting from the addition of muramyl tripeptide encapsulated in liposomes to a HDMTX, PDD, isofosfamide and doxorubicin regimen [232]. The authors observed that the addition of tripeptide, in practice, did not enhance event-free survival. Crews et al. demonstrated the relation of MTX pharmacokinetics between patient age and outcome in different combination therapies of osteosarcoma [13]. After the HDMTX administration, higher mean serum concentrations, higher exposures, and lower mean clearance of MTX were associated with poorer therapy outcomes. The safety and efficacy of osteosarcoma therapy conducted with HDMTX in adolescents compared with young adults were tested by Wippel et al. [275]. The obtained results showed that young adult patients (aged 19–38) are more likely to experience a delay in MTX clearance when compared to adolescents (aged 7–18) despite using the similar supportive rescue. Application of HDMTX in osteosarcoma neoadjuvant chemotherapy used in the frame of a randomized cooperative trial is presented in the work of Winkler et al. [233]. Two groups of patients were treated with two approaches: the first one was based on preoperative chemotherapy containing HDMTX applied with the triple drug combination of bleomycin, CTX and dactinomycin (BCD) and postoperative salvage treatment with doxorubicin and PDD, and the second, preoperative chemotherapy with doxorubicin-PDD or doxorubicin-BCD system application and postoperative treatment with ifosfamide. The histologic response rate was significantly lower in the case of preoperative chemotherapy containing HDMTX (first approach); however, the four-year metastasis-free survival rate was improved in the case of using doxorubicin-PDD in preoperative chemotherapy (the second approach).
HDMTX-based neoadjuvant chemotherapy for localized osteosarcoma treatment has been described by Bacci et al. [234,235]. The obtained results showed that application of aggressive neoadjuvant chemotherapy: HDMTX i.v. followed by PDD and doxorubicin was allowed to carry more than 60% of non-metastatic osteosarcoma to the extremities, and in about of 80% of patients it allowed the avoidance of an amputation. Ifosfamide and etoposide seem to be indicated in patients who did not respond to preoperative chemotherapy good enough. Combination of MTX and doxorubicin, described by Geller et al. [236], ensured disease-free survival in about 60% of patients. Frequently used in the past combination of bleomycin, CTX and dactinomycin (BCD) has been replaced by a regimen of MTX and Adriamycin. Systemic chemotherapy consisting of HDMTX, PDD, and doxorubicin allowed a cure in approximately 70% of patients with localized osteosarcoma. Unfortunately, a long-term survival rate in patients with metastases was here less than 20%. A new combination containing HDMTX and trimetrexate (a structural analogue of MTX) was tested in the treatment of patients with recurrent osteosarcoma. Another new regimen mentioned in this work was the set containing zoledronic acid in combination with HDMTX, PDD, doxorubicin, ifosfamide, and etoposide, especially indicated for patients with newly diagnosed metastatic osteosarcoma. In the work, the mechanisms responsible for MTX resistance were presented. MTX resistance in osteosarcoma treatment and the ways it is overcome are discussed also in papers of Guo et al. [276], Wang et al. [277] and Xu et al. [278]. Xu et al. proposed a relatively novel method to overcome the MTX resistance in osteosarcoma. The authors used the capacity of microRNAs (miRNAs) to regulate diverse biological processes, including drug resistance. miRNAs are small non-coding RNAs implicated in a growing number of human diseases, including osteosarcoma [279]. Based on obtained results Xu et al. determined that miR-29 family can inhibit MTX resistance and induce cell apoptosis in osteosarcoma. In the end, targeting the miR-29 family might provide to overcome the HDMTX-induced cytotoxicity in osteosarcoma treatment.

3.2.8. Methotrexate-Based Therapy of Gestational Trophoblastic Disease

Gestational trophoblastic neoplasia (GTN) is a pregnancy-related cancer. These tumours are formed when the cells of the womb in the process of placenta formation begin to proliferate in an uncontrolled way. The most invasive neoplasm belonging to this group cancer is choriocarcinoma characterized with quickly haematogenous spreading (mainly to the lungs, brain and liver), but also with high sensitivity to chemotherapy and a very good prognosis.
In the work of Abräo et al., three treatment regimens—MTX, dactinomycin (ACT) and the combination of MTX and ACT (MACT) as first-line chemotherapy in GTN—are described and compared [237]. The determined rates of total remission were 69%, 61% and 79%, respectively, in the MTX, ACT and MACT regimens, and the adverse reaction rates 29%, 19% and 63% in MTX, ACT and MACT, respectively. As one can see, the most effective therapeutic effect was obtained in the MACT scheme, but in this case the toxic side effects were also the highest. The most optimal agent tested under these studies seems to be ACT. Lurain et al. analysed the efficacy and toxicity of MTX-based chemotherapy in patients with non-metastatic gestational trophoblastic disease [280]. The subsequent stages of therapy included MTX monotherapy, ACT monotherapy, combined chemotherapy and finally hysterectomy. The therapy was well tolerated (the most common side effect was severe stomatitis) and brought healing to all the patients. Based on study results, the authors determined that high human chorionic gonadotropin (hCG) levels are a major factor associated with MTX resistance development. The same GTN therapy profile was applied in the studies described by Chapman-Davis et al. [281], similar to that of Lurain et al. [280]. The deep analysis of single-agent MTX-based chemotherapies performed on nearly 1000 patients are described in the work of Couder et al. [282]. The studies included the tumour treatments in patients with low-risk GTN, who, according to the FIGO classification, were indicated for MTX chemotherapy. Based on the analysed data, the authors concluded that the number of chemotherapy courses needed to normalize the hCG level may be a predictive factor of tumour recurrence—in the patients needing more than four MTX courses, the risk of relapse was definitely higher. The paper of Lawrie et al. [283] presents results of seven randomised controlled trials performed in large group of patients with low risk GTN. Each group consisted of about 100 patients, and in chemotherapy procedures single agent MTX or ACT were applied (both drugs in different doses and in different time intervals were administrated). In general, both therapies were highly effective and well tolerated; however, the authors suggested a slight therapeutic advantage of ACT chemotherapy over the MTX one. Two therapies applied in patients with high-risk GTN are described in the work of Han et al. [238]. The therapies were conducted using the following regimens: HDMTX-etoposide and etoposide-cisplatin in combination with etoposide-MTX-dactinomycin (EP-EMA). The first preliminary studies performed by these researchers showed a better toxicity profile of HDMTX-etoposide than EP-EMA. According to the authors, the HDMTX-etoposide regimen can be indicated for patients with high-risk GTN. The works of May et al., Aminimoghaddam et al. and Alazzam et al. present chemotherapeutic management of women with all stages of GTN as well as with recurrent disease [284,285,286]. In these papers, besides the information about monotherapies conducted with single drugs of, for example, MTX, dactinomycin, etoposide, cisplatin, there are also described application and effectiveness of different regimens. Based on the study results, the authors indicate MTX or ACT as first-line single drugs for low-risk GTN treating and etoposide, MTX, dactinomycin, CTX and vincristine as first-line regimen for high-risk GTN therapy. Nevertheless, the choice of therapy must always take into account the patient’s physical condition, drug tolerance, drug resistance, adverse reactions and the stage of the tumour. The article of Skubisz et al. presents MTX and its mechanism of action, as well its application in GTN and ectopic pregnancy treatment [287]. The authors classify chemotherapy of patients with GTN into two groups, of low risk and high risk, depending on Prognostic Index Score (PIS) determined by WHO. Low-risk women are highly likely to respond to single-agent chemotherapy with MTX or dactinomycin. MTX followed by LV is a first-line treatment in women with low risk GTN. High-risk GTN is defined as stage IV disease or stage II-III disease with a PIS index of seven or higher and in these cases multiagent chemotherapy is needed. Combination regimen of etoposide, MTX, dactinomycin, CTX and vincristine is the first-line regimen for high risk GTN and it is a relatively well-tolerated regimen. The work of Agarval et al. concerns the validation of the uterine artery pulsatility index (UAPI, a measure of tumour vascularity) as a useful indicator for predicting resistance to MTX chemotherapy [288]. A similar issue is also the subject of the works of Secki et al. [289] and Hussain et al. [290]. Stratification of patients of low- or high-risk GTN for single-agent chemotherapy (with MTX-LV or ACT) or multi-agents chemotherapy, respectively, is based on the assessment of the CXH and FIGO indexes. However, this evaluation does not take into account the effect of MTX resistance on the effectiveness of therapy, which is very important especially in the case of low-risk GTN patients indicated for MTX-LV therapy [291,292]. The authors’ findings showed that the UAPI index can be a non-invasive marker of tumour vascularity and MTX resistance, and a useful tool for the indication of appropriate chemotherapy. A low UAPI index value indicates for tumour formation, and increasing the UAPI index value in patients with GTN indicates emerging MTX resistance.

4. Conclusions

In the present review, we take an overview of present state knowledge as regards to MTX, a structural analogue of FA, which is one of the most effective and extensively used drugs for treating severe and resistant forms of neurological and autoimmune diseases or many kinds of cancers. We have carefully overviewed the scientific literature (articles, trial studies and guidelines by FDA and EMA) to describe MTX’s action mechanisms and its applications. MTX is already a well-known DMARD and chemotherapy agent. It has been used in low doses as an effective drug in RA for many years and in high doses in the therapy of various types of cancer for decades.
Application of MTX in high doses in cancer therapies is associated usually with serious side effects, even life-threatening for patients. However, prolonged use of MTX in low doses can also result in occurrence of serious side effects. There are numerous reports in the literature concerning MTX-induced neoplasms (mainly lymphomas [293,294,295,296,297,298] and lung cancer [271,299], but also breast cancer [268], hepatocarcinoma [300], acute leukemia [301]) in patients with RA or psoriasis treated with LDMTX administered continuously and gradually for a long period of time.
Relatively recently, reports have appeared in the literature about the application of MTX in the treatment of neurological diseases MG [302] and AD (as a drug potentially able to reduce the risk of dementia) [103,104,105,106]. However, these medical experiments have been conducted on small population sizes and, up to now, without significant results, and more detailed studies are needed for determining the conclusive effect of MTX with regards to these diseases.
Nevertheless, based on the literature data demonstrated in our review, one can conclude that MTX therapy provides sustained clinical benefits with controlled and minimized side effects (using recommended rescue), even in off-label applications. In summary, MTX has been used over 60 years to treat variety of autoimmune disorders with the same or greater efficacy than most other synthetic DMARDs, e.g., AZT, and is also used alone or in combination with other anticancer agents in different types of cancer treatments and still remains of great interest to researchers all over the world.

Author Contributions

Conceptualization, P.K., P.K.H. and E.G.; writing—original draft preparation, P.K., P.K.H. and E.G.; writing—review and editing, P.K., P.K.H., R.C. and E.G.; visualization, P.K.H.; supervision, E.G.; project administration, P.K.; All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by statutory activity of the Institute of Nuclear Chemistry and Technology, Warsaw, Poland. The contribution of Paweł Krzysztof Halik has been done in the frame of the National Centre for Research and Development Project No POWR.03.02.00-00-I009/17 (Radiopharmaceuticals for molecularly targeted diagnosis and therapy, RadFarm, Operational Project Knowledge Education Development 2014-2020 co-financed by European Social Fund). The contribution of Raphael Chebosion Chesori was realized within the framework of the International Atomic Energy Agency Technical Cooperation Fellowship FS-RAF0052-1805593.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

5-FU5-fluorouracil
A1, A2A, A2B, A3Adenosine receptors
A549Human non-small cell lung carcinoma cell line
ACTDactinomycin, actinomycin D
ADAlzheimer’s disease
ADAAdenosine deaminase
AICART5-Aminoimidazole-4-carboxamide ribonucleotide transformylase
AICAR5-Aminoimidazole-4-carboxamide ribonucleotide
ALLAcute lymphoblastic leukaemia
AMLAcute myelogenous leukaemia
AMPAdenosine monophosphate
AMPDAAdenosine monophosphate deaminase
Ara-CCytarabine, cytosine arabinoside
ASAAcetylsalicylic acid, aspirin
ATCVAnthracyclines-taxanes-capecitabine-vinorelin
ATPAdenosine triphosphate
AZTAzathioprine
BBBBlood-brain barrier
BCDBleomycin-cyclophosphamide-dactinomycin
C225Cetuximab
cAMPCyclic adenosine monophosphate
CL1-0Human non-small cell lung carcinoma cell line
CD73ecto-5′-nucleotidase
CLLChronic lymphocytic leukaemia
CMFCyclophosphamide-methotrexate-5-fluorouracil
CMFEVCyclophosphamide-methotrexate-5-fluorouracil-epirubicin or vincristine
CMLChronic myelogenous leukaemia
CMVCisplatin-methotrexate-vinblastine
CNSCentral nervous system
COBCisplatin-Oncovin(vincristine)-bleomycin
CXHChering Cross Hospital
DFSDisease-free survival
DHFRDihydrofolate reductase
DIACisplatin-ifosfamide-doxorubicin
DMARDs Disease-modifying anti-rheumatic drugs
DNA Deoxyribonucleic acid
dUMPDeoxyuridine monophosphate
EMAEuropean Medicines Agency
EP-EMAEtoposide-cisplatin + etoposiede-methotrexate-dactinomycin
EPREpirubicin
EROestrogen receptor
FAFolic acid
FAC 5-Fluorouracil-Adriamycin-cyclophosphamide
FAICAR5-Formamidoimidazole- 4-carboxamide ribotide
FIGOThe International Federation of Gynecology and Obstetrics
FDAThe US Food and Drug Administration
FRsFolate receptors
FPGSFolylpolyglutamyl synthase
G5Generation 5
GCSGlucocorticosteroid
Glu Glutamic acid
GMPGuanosine monophosphate
GPIGlycosyl-phosphatidyl-inositol
GTNGestational trophoblastic neoplasia
hCGHuman chorionic gonadotropin
HDMTXHigh-dose methotrexate
HDMVACHigh-dose methotrexate-vinblastine-Adriamycin-cisplatin
HER2Human epidermal growth factor receptor-2
HNSCCHead and neck squamous cell carcinoma
HSAHuman serum albumin
IDMTXIntermediate-dose methotrexate
IFN-γInterferon γ
IL Interleukin
IMPInosine monophosphate
i.v.Intravenous
LDMTXLow-dose methotrexate
LLCMurine epidermal lung carcinoma cell line
LVLeucovorin, folinic acid
MACTMethotrexate and dactinomycin
MAPMethotrexate-doxorubicin-cisplatin
MFMycosis fungoides
MGMyasthenia gravis
miRNAmicroRNA
MLCCMethotrexate-leucovorin-cyclophosphamide-cytosine arabinoside
MMMitomycin C and methotrexate
MNPMitomycin C-vinorelbine-cisplatinum
M-MNPMethotrexate-mitomycin C-vinorelbine-cisplatinum
M-MVPMethotrexate-mitomycin C-vinblastin-cisplatin
MOACMethotrexate-vincristine(Oncovin)-doxorubicin(Adriamycin)-cyclophosphamide
MSMultiple sclerosis
MTXMethotrexate
MULMethotrexate-tegafur-uracil-leucovorin
MVACMethotrexate-vinblastine-Adriamycin-cisplatin
MVPMitomycin C-vinblastin-cisplatin
NHLNon-Hodgkin lymphoma
NPNanoparticulate
NSAIDsNon-steroidal anti-inflammatory drugs
NSCLCNon-small cell lung cancer
ORObjective response
OSOverall survival
PABAp-Aminobenzoic acid
PAMAMpoly(Amidoamine)
PCFTProton-coupled folate transporter
PCNPsPEGylated chitosan nanoparticles
PCNSLPrimary central nervous system lymphoma
PDDCisplatin, cis-diammindichloridoplatin
PEGpoly(Ethylene glycol)
PEPEpolyester-co-polyether
PFSProgression-free survival
PISPrognostic index score
PLApoly(lactic acid)
PLGApoly(d,l-lactide-co-glycolide)
PMXPemetrexed
PRProgesterone receptor
RARheumatoid arthritis
R-CHOPRituximab-cyclophosphamide-doxorubicin-vincristine-prednisolone
RFC1Reduced folate carrier 1
R-MADRituximab-methotrexate-cytarabine-dexamethasone
R-MPV Rituximab-methotrexate-procarbazine-vincristine
RNARibonucleic acid
SCLCSmall cell lung cancer
S-SLMRat osteosarcoma cell line
TCCTransitional cell carcinoma
THFTetrahydrofolate
TNBCTriple-negative breast cancer
TNF-αTumour necrosis factor α
TYMSThymidylate synthase
UAPIUterine artery pulsatility index
UCUrothelial cancer
WHOWorld Health Organization

References

  1. Farber, S. Some observations on the effect of folic acid antagonists on acute leukemia and other forms of incurable cancer. Blood 1949, 4, 160–167. [Google Scholar] [CrossRef] [Green Version]
  2. Gubner, R.; August, S.; Ginsberg, V. Therapeutic suppression of tissue reactivity. II. Effect of aminopterin in rheumatoid arthritis and psoriasis. Am. J. Med. Sci. 1951, 221, 176–182. [Google Scholar] [CrossRef]
  3. Edmundson, W.F.; Guy, W.B. Treatment of Psoriasis with Folic Acid Antagonists. AMA Arch. Derm. 1958, 78, 200–203. [Google Scholar] [CrossRef] [PubMed]
  4. Williams, H.J.; Willkens, R.F.; Samuelson, C.O., Jr.; Alarcon, G.S.; Guttadauria, M.; Yarboro, C.; Polisson, R.P.; Weiner, S.R.; Luggen, M.E.; Billingsley, L.M.; et al. Comparison of low-dose oral pulse methotrexate and placebo in the treatment of rheumatoid arthritis. A controlled clinical trial. Arthritis Rheum. 1985, 28, 721–730. [Google Scholar] [CrossRef]
  5. Weinstein, A.; Marlowe, S.; Korn, J.; Farouhar, F. Low-dose methotrexate treatment of rheumatoid arthritis. Long-term observations. Am. J. Med. 1985, 79, 331–337. [Google Scholar] [CrossRef] [PubMed]
  6. Methotrexate Injection, USP. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/011719s125lbl.pdf (accessed on 2 January 2020).
  7. Methotrexate oral solution, FDA Full Prescribing Information. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/208004s002lbl.pdf (accessed on 2 January 2020).
  8. Frei, E., III; Jaffe, N.; Tattersall, M.H.N.; Pitman, S.; Parker, L. New Approaches to Cancer Chemotherapy with Methotrexate. N. Engl. J. Med. 1975, 292, 846–851. [Google Scholar] [CrossRef] [PubMed]
  9. Evans, W.E.; Crom, W.R.; Abromowitch, M.; Dodge, R.; Look, A.T.; Bowman, W.P.; George, S.L.; Pui, C.H. Clinical Pharmacodynamics of High-Dose Methotrexate in Acute Lymphocytic Leukemia. N. Engl. J. Med. 1986, 314, 471–477. [Google Scholar] [CrossRef] [PubMed]
  10. Natale, R.B.; Yagoda, A.; Watson, R.C.; Whitmore, W.F.; Blumenreich, M.; Braun, D.W., Jr. Methotrexate: An Active Drug in Bladder Cancer. Cancer 1981, 47, 1246–1250. [Google Scholar] [CrossRef]
  11. Huennekens, F.M. The methotrexate story: A paradigm for development of cancer chemotherapeutic agents. Advan. Enzym. Regul. 1994, 34, 397–419. [Google Scholar] [CrossRef]
  12. Bonadonna, G.; Valagussa, P.; Moliterni, A.; Zambetti, M.; Brambilla, C. Adjuvant Cyclophosphamide, Methotrexate, and Fluorouracil in Node-Positive Breast Cancer—The Results of 20 Years of Follow-up. N. Engl. J. Med. 1995, 332, 901–906. [Google Scholar] [CrossRef]
  13. Crews, K.R.; Liu, T.; Rodriguez-Galindo, C.; Tan, M.; Meyer, W.H.; Panetta, J.C.; Link, M.P.; Daw, N.C. High-dose methotrexate pharmacokinetics and outcome of children and young adults with osteosarcoma. Cancer 2004, 100, 1724–1733. [Google Scholar] [CrossRef] [PubMed]
  14. Tugwell, P.; Bennett, K.; Gent, M. Methotrexate in rheumatoid arthritis. Health and Public Policy Committee, American College of Physicians. Ann. Intern. Med. 1987, 107, 418–419. [Google Scholar]
  15. Cronstein, B.N. Low-Dose Methotrexate: A Mainstay in the Treatment of Rheumatoid Arthritis. Pharmacol. Rev. 2005, 57, 163–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Roenigk, H.H., Jr.; Auerbach, R.; Maibach, H.I.; Weinstein, G.D. Methotrexate in psoriasis: Revised guidelines. J. Am. Acad. Dermatol. 1988, 19, 145–156. [Google Scholar] [CrossRef]
  17. Karaahmet, O.Z.; Bal, A.; Dulgeroglu, D.; Bahceci, H.K.; Cakci, A. Methotrexate treatment in myasthenia gravis. J. Clin. Neuromuscul. Dis. 2014, 16, 106–107. [Google Scholar] [CrossRef]
  18. Lémann, M.; Zenjari, T.; Bouhnik, Y.; Cosnes, J.; Mesnard, B.; Rambaud, J.C.; Modigliani, R.; Cortot, A.; Colombel, J.F. Methotrexate in Crohn’s disease: Long-term efficacy and toxicity. Am. J. Gastroenterol. 2000, 95, 1730–1734. [Google Scholar] [CrossRef]
  19. Goodkin, D.E.; Rudick, R.A.; VanderBrug Medendorp, S.; Daughtry, M.M.; Schwetz, K.M.; Fischer, J.; Van Dyke, C. Low-dose (7.5 mg) oral methotrexate reduces the rate of progression in chronic progressive multiple sclerosis. Ann. Neurol. 1995, 37, 30–40. [Google Scholar] [CrossRef]
  20. ACEP Clinical Policies Committee and the Clinical Policies Subcommittee on Early Pregnancy. Clinical policy: Critical issues in the initial evaluation and management of patients presenting to the emergency department in early pregnancy. Ann. Emerg. Med. 2003, 41, 123–133. [Google Scholar] [CrossRef]
  21. Jacobs, J.W. Optimal use of non-biologic therapy in the treatment of rheumatoid arthritis. Rheumatology (Oxford) 2012, 51 (Suppl. S4), iv3–iv8. [Google Scholar] [CrossRef] [Green Version]
  22. Singh, J.A.; Furst, D.E.; Bharat, A.; Curtis, J.R.; Kavanaugh, A.F.; Kremer, J.M.; Moreland, L.W.; O’Dell, J.; Winthrop, K.L.; Beukelman, T.; et al. 2012 update of the 2008 American College of Rheumatology recommendations for the use of disease-modifying antirheumatic drugs and biologic agents in the treatment of rheumatoid arthritis. Arthritis Care Res. (Hoboken) 2012, 64, 625–639. [Google Scholar] [CrossRef] [Green Version]
  23. Chattopadhyay, S.; Moran, R.G.; Goldman, I.D. Pemetrexed: Biochemical and cellular pharmacology, mechanisms, and clinical applications. Mol. Cancer. Ther. 2007, 6, 404–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Matera, C.; Gomila, A.M.J.; Camarero, N.; Libergoli, M.; Soler, C.; Gorostiza, P. Photoswitchable Antimetabolite for Targeted Photoactivated Chemotherapy. J. Am. Chem. Soc. 2018, 140, 15764–15773. [Google Scholar] [CrossRef] [Green Version]
  25. Raimondi, M.V.; Randazzo, O.; La Franca, M.; Barone, G.; Vignoni, E.; Rossi, D.; Collina, S. DHFR Inhibitors: Reading the Past for Discovering Novel Anticancer Agents. Molecules 2019, 24, 1140. [Google Scholar] [CrossRef] [Green Version]
  26. Huschtascha, L.I.; Bartier, W.A.; Ross, C.E.; Tattersall, M.H.N. Characteristics of cancer cell death after exposure to cytotoxic drugs in vitro. Br. J. Cancer 1996, 73, 54–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Chan, E.S.L.; Cronstein, B.N. Mechanism of Action of Methotrexate. Bull. Hosp. Jt. Dis. 2013, 71 (Suppl. S1), S5–S8. [Google Scholar]
  28. Genestier, L.; Paillot, R.; Quemeneur, L.; Izeradjene, K.; Revillard, J.P. Mechanisms of action of methotrexate. Immunopharmacology 2000, 47, 247–257. [Google Scholar] [CrossRef]
  29. Tian, H.; Cronstein, B.N. Understanding the mechanisms of action of methotrexate: Implications for the treatment of rheumatoid arthritis. Bull. NYU Hosp. Jt. Dis. 2007, 65, 168–173. [Google Scholar] [PubMed]
  30. Inoue, K.; Yuasa, H. Molecular Basis for Pharmacokinetics and Pharmacodynamics of Methotrexate in Rheumatoid Arthritis Treatment. Drug Metab. Pharmacokinet. 2014, 29, 12–19. [Google Scholar] [CrossRef] [Green Version]
  31. Matherly, L.H.; Goldman, D.I. Membrane transport of folates. In Vitamins and Hormones; Litwack, G., Ed.; Elsevier: Amsterdam, The Netherlands, 2003; Volume 66, pp. 403–456. [Google Scholar] [CrossRef]
  32. Sabharanjak, S.; Mayor, S. Folate receptor endocytosis and trafficking. Adv. Drug. Deliv. Rev. 2004, 56, 1099–1109. [Google Scholar] [CrossRef]
  33. Nogueira, E.; Sarria, M.P.; Azoia, N.G.; Antunes, E.; Loureiro, A.; Guimaraes, D.; Noro, J.; Rollett, A.; Guebitz, G.; Cavaco-Paulo, A. Internalization of Methotrexate Conjugates by Folate Receptor-α. Biochemistry 2018, 57, 6780–6786. [Google Scholar] [CrossRef] [Green Version]
  34. Baugh, C.M.; Krumdieck, C.L.; Nair, M.G. Polygammaglutamyl metabolites of methotrexate. Biochem. Biophys. Res. Commun. 1973, 52, 27–34. [Google Scholar] [CrossRef]
  35. Dervieux, T.; Furst, D.; Lein, D.O.; Capps, R.; Smith, K.; Walsh, M.; Kremer, J. Polyglutamation of methotrexate with common polymorphisms in reduced folate carrier, aminoimidazole carboxamide ribonucleotide transformylase, and thymidylate synthase are associated with methotrexate effects in rheumatoid arthritis. Arthritis Rheum. 2004, 50, 2766–2774. [Google Scholar] [CrossRef] [PubMed]
  36. Chabner, B.A.; Allegra, C.J.; Curt, G.A.; Clendeninn, N.J.; Baram, J.; Koizumi, S.; Drake, J.C.; Jolivet, J. Polyglutamation of methotrexate: Is methotrexate a prodrug? J. Clin. Invest. 1985, 76, 907–912. [Google Scholar] [CrossRef] [PubMed]
  37. Wu, K.M. A New Classification of Prodrugs: Regulatory Perspectives. Pharmaceuticals 2009, 2, 77–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Allegra, C.J.; Chabner, B.A.; Drake, J.C.; Lutz, R.; Rodbard, D.; Jolivet, J. Enhanced inhibition of thymidylate synthase by methotrexate polyglutamates. J. Biol. Chem. 1985, 260, 9720–9726. [Google Scholar]
  39. Baggott, J.E.; Vaughn, W.H.; Hudson, B.B. Inhibition of 5-aminoimidazole4-carboxamide ribotide transformylase, adenosine deaminase and 5’-adenylate deaminase by polyglutamates of methotrexate and oxidized folates and by 5-aminoimidazole-4-carboxamide riboside and ribotide. Biochem. J. 1986, 236, 193–200. [Google Scholar] [CrossRef] [Green Version]
  40. Allegra, C.J.; Drake, J.C.; Jolivet, J.; Chabner, B.A. Inhibition of phosphoribosylaminoimidazolecarboxamide transformylase by methotrexate and dihydrofolic acid polyglutamates. Proc. Natl. Acad. Sci. USA 1985, 82, 4881–4885. [Google Scholar] [CrossRef] [Green Version]
  41. Sant, M.E.; Lyons, S.D.; Phillips, L.; Christopherson, R.I. Antifolates Induce Inhibition of Amido Phosphoribosyltransferase in Leukemia. Cell J. Biol. Chem. 1992, 267, 11038–11045. [Google Scholar]
  42. Fairbanks, L.D.; Rückermann, K.; Qiu, Y.; Hawrylowicz, C.M.; Richards, D.F.; Swaminathan, R.; Kirschbaum, B.; Simmonds, H.A. Methotrexate Inhibits the First Committed Step of Purine Biosynthesis in Mitogen-Stimulated Human T-lymphocytes: A Metabolic Basis for Efficacy in Rheumatoid Arthritis? Biochem. J. 1999, 341, 143–152. [Google Scholar] [CrossRef]
  43. Genestier, L.; Paillot, R.; Fournel, S.; Ferraro, C.; Miossec, P.; Revillard, J.P. Immunosuppressive Properties of Methotrexate: Apoptosis and Clonal Deletion of Activated Peripheral T Cells. J. Clin. Invest. 1998, 102, 322–328. [Google Scholar] [CrossRef]
  44. Morabito, L.; Montesinos, M.C.; Schreibman, D.M.; Balter, L.; Thompson, L.F.; Resta, R.; Carlin, G.; Huie, M.A.; Cronstein, B.N. Methotrexate and Sulfasalazine Promote Adenosine Release by a Mechanism that Requires Ecto-59-nucleotidase–mediated Conversion of Adenine Nucleotides. J. Clin. Invest. 1998, 101, 295–300. [Google Scholar] [CrossRef] [PubMed]
  45. Montesinos, M.C.; Desai, A.; Delano, D.; Chen, J.F.; Fink, J.S.; Jacobson, M.A.; Cronstein, B.N. Adenosine A2A or A3 receptors are required for inhibition of inflammation by methotrexate and its analog MX-68. Arthritis Rheum. 2003, 48, 240–247. [Google Scholar] [CrossRef] [PubMed]
  46. Varani, K.; Vincenzi, F.; Tosi, A.; Targa, M.; Masieri, F.F.; Ongaro, A.; De Mattei, M.; Massari, L.; Borea, P.A. Expression and Functional Role of Adenosine Receptors in Regulating Inflammatory Responses in Human Synoviocytes. Br. J. Pharmacol. 2010, 160, 101–115. [Google Scholar] [CrossRef] [Green Version]
  47. Stamp, L.K.; Hazlett, J.; Roberts, R.L.; Frampton, C.; Highton, J.; Hessian, P.A. Adenosine receptor expression in rheumatoid synovium: A basis for methotrexate action. Arthritis Res. Ther. 2012, 14, R138. [Google Scholar] [CrossRef] [Green Version]
  48. Vincenzi, F.; Padovan, M.; Targa, M.; Corciulo, C.; Giacuzzo, S.; Merighi, S.; Gessi, S.; Govoni, M.; Borea, P.A.; Varani, K. A2A Adenosine Receptors Are Differentially Modulated by Pharmacological Treatments in Rheumatoid Arthritis Patients and Their Stimulation Ameliorates Adjuvant-Induced Arthritis in Rats. PLoS ONE 2013, 8, e54195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Cronstein, B.N.; Eberle, M.A.; Gruber, H.E.; Levin, R.I. Methotrexate inhibits neutrophil function by stimulating adenosine release from connective tissue cells. Proc. Natl. Acad. Sci. USA 1991, 88, 2441–2445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Cronstein, B.N.; Levin, R.I.; Belanoff, J.; Weissmann, G.; Hirschhorn, R. Adenosine: An endogenous inhibitor of neutrophil-mediated injury to endothelial cells. J. Clin. Invest. 2001, 78, 760–770. [Google Scholar] [CrossRef]
  51. Leroux, J.L.; Damon, M.; Chavis, C.; Crates de Paulet, A.; Blotman, F. Effects of methotrexate on leukotriene and derivated lipoxygenase synthesis in polynuclear neutrophils in rheumatoid polyarthritis. Rev. Rheum. Mal. Osteoartic. 1992, 59, 587–591. [Google Scholar]
  52. Brody, M.; Bohm, I.; Bauer, R. Mechanism of action of methotrexate: Experimental evidence that methotrexate blocks the binding of interleukin-1 beta to the interleukin-1 receptor on target cells. Eur. J. Chem. Clin. Biochem. 1993, 31, 667–674. [Google Scholar] [CrossRef] [Green Version]
  53. Bouma, M.G.; Stad, R.K.; van der Wildenberg, F.A.J.M.; Buurman, W.A. Differential regulatory effects of adenosine on cytokine release by activated human monocytes. J. Immunol. 1994, 153, 4159–4168. [Google Scholar]
  54. Becker, C.; Barbulescu, K.; Hildner, K.; Meyer zum Buschenfelde, K.H.; Neurath, M.F. Activation and methotrexate-mediated suppression of the TNF alpha promoter in T cells and macrophages. Ann. N. Y. Acad. Sci. 1998, 859, 311–314. [Google Scholar] [CrossRef] [PubMed]
  55. Constantin, A.; Loubet-Lescoulie, P.; Lambert, N.; Yassine-Diab, B.; Abbal, M.; Mazieres, B.; de Preval, C.; Cantagrel, A. Antiinflammatory and immunoregulatory action of methotrexate in the treatment of rheumatoid arthritis: Evidence of increased interleukin-4 and interleukin-10 gene expression demonstrated in vitro by competitive reverse transcriptase-polymerase chain reaction. Arthritis Rheum. 1998, 41, 48–57. [Google Scholar] [CrossRef] [PubMed]
  56. Hasko, G.; Cronstein, B.N. Adenosine: An endogenous regulator of innate immunity. Trends Immunol. 2004, 25, 33–39. [Google Scholar] [CrossRef] [PubMed]
  57. Bouma, M.G.; van der Wildenberg, F.A.; Buurman, W.A. Adenosine inhibits cytokine release and expression of adhesion molecules by activated human endothelial cells. Am. J. Physiol. 1996, 270, C522–C529. [Google Scholar] [CrossRef] [PubMed]
  58. Nesher, G.; Moore, T.L. The in vitro effects of methotrexate on peripheral blood mononuclear cells. Modulation by methyl donors and spermidine. Arthritis Rheum. 1990, 33, 954–959. [Google Scholar] [CrossRef]
  59. Yukioka, K.; Wakitani, S.; Yukioka, M.; Furumitsu, Y.; Shichikawa, K.; Ochi, T.; Goto, H.; Matsui-Yuasa, I.; Otani, S.; Nishizawa, Y. Polyamine levels in synovial tissues and synovial fluids of patients with rheumatoid arthritis. J. Rheumatol. 1992, 19, 689–692. [Google Scholar]
  60. Nesher, G.; Osborn, T.G.; Moore, T.L. In vitro effects of methotrexate on polyamine levels in lymphocytes from rheumatoid arthritis patients. Clin. Exp. Rheumatol. 1996, 14, 395–399. [Google Scholar]
  61. Nesher, G.; Osborn, T.G.; Moore, T.L. Effect of treatment with methotrexate, hydroxychloroquine, and prednisone on lymphocyte polyamine levels in rheumatoid arthritis: Correlation with the clinical response and rheumatoid factor synthesis. Clin. Exp. Rheumatol. 1997, 15, 343–347. [Google Scholar]
  62. Wong, P.T.; Choi, S.K. Mechanisms and Implications of Dual-Acting Methotrexate in Folate-Targeted Nanotherapeutic Delivery. Int. J. Mol. Sci. 2015, 16, 1772–1790. [Google Scholar] [CrossRef] [Green Version]
  63. Theti, D.S.; Jackman, A.L. The Role of α-Folate Receptor-Mediated Transport in the Antitumor Activity of Antifolate Drugs. Clin. Cancer Res. 2004, 10, 1080–1089. [Google Scholar] [CrossRef] [Green Version]
  64. Kerschbaumer, A.; Sepriano, A.; Smolen, J.S.; van der Heijde, D.; Dougados, M.; van Vollenhoven, R.; McInnes, I.B.; Bijlsma, J.W.J.; Burmester, G.R.; de Wit, M.; et al. Efficacy of pharmacological treatment in rheumatoid arthritis: A systematic literature research informing the 2019 update of the EULAR recommendations for management of rheumatoid arthritis. Ann. Rheum. Dis. 2020, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Smolen, J.S.; Aletaha, D.; Bijlsma, J.W.; Breedveld, F.C.; Boumpas, D.; Burmester, G.; Combe, B.; Cutolo, M.; de Wit, M.; Dougados, M.; et al. Treating rheumatoid arthritis to target: Recommendations of an international task force. Ann. Rheum. Dis. 2010, 69, 631–637. [Google Scholar] [CrossRef] [PubMed]
  66. Díaz-Manera, J.; García, R.R.; Illa, I. Treatment strategies for myasthenia gravis: An update. Expert Opin. Pharmacother. 2012, 13, 1873–1883. [Google Scholar] [CrossRef] [PubMed]
  67. Gotterer, L.; Li, Y. Maintenance immunosuppression in myasthenia gravis. J. Neurol. Sci. 2016, 369, 294–302. [Google Scholar] [CrossRef] [PubMed]
  68. Chantam, W. Rheumatic manifestations of systemic disease: Sarcoidosis. Curr. Opin. Rheumatol. 2010, 22, 85–90. [Google Scholar] [CrossRef]
  69. Baughman, R.P.; Lower, E.E. A clinical approach to the use of methotrexate for sarcoidosis. Thorax 1999, 54, 742–746. [Google Scholar] [CrossRef] [Green Version]
  70. Mahr, A.D.; Jover, J.A.; Spiera, R.F.; Hernández-García, C.; Fernández-Gutiérrez, B.; LaValley, M.P.; Merkel, P.A. Adjunctive Methotrexate for Treatment of Giant Cell Arteritis. An Individual Patient Data Meta-Analysis. Arthritis Rheum. 2007, 56, 2789–2797. [Google Scholar] [CrossRef]
  71. Ferrara, G.; Mastrangelo, G.; Barone, P.; La Torre, F.; Martino, S.; Pappagallo, G.; Ravelli, A.; Taddio, A.; Zulian, F.; Cimaz, R. Methotrexate in juvenile idiopathic arthritis: Advice and recommendations from the MARAJIA expert consensus meeting. Pediatr. Rheumatol. 2018, 16, 46. [Google Scholar] [CrossRef] [Green Version]
  72. Joshi, P.; Dhaneshwar, S.S. An update on Disease Modifying Antirheumatic drugs. Inflamm. Allergy Drug Target 2014, 13, 249–261. [Google Scholar] [CrossRef]
  73. Gilhus, N.E. Myasthenia and the neuromuscular junction. Curr. Opin. Neurol. 2012, 25, 523–529. [Google Scholar] [CrossRef]
  74. Andersen, J.B.; Heldal, A.T.; Engeland, A.; Gilhus, N.E. Myasthenia gravis epidemiology in a national cohort; combining multiple disease registries. Acta Neurol. Scand. 2014, 129, 26–31. [Google Scholar] [CrossRef]
  75. Gilhus, N.E.; Hong, Y. Maternal myasthenia gravis represents a risk for the child through autoantibody transfer, immunosuppressive therapy and genetic influence. Eur. J. Neurol. 2018, 25, 1402–1409. [Google Scholar] [CrossRef] [PubMed]
  76. Kupersmith, M.J.; Latkany, R.; Homel, P. Development of generalized disease at 2 years in patients with ocular myasthenia gravis. Arch. Neurol. 2003, 60, 243–248. [Google Scholar] [CrossRef] [PubMed]
  77. Sathasivam, S. Steroids and immunosuppressant drugs in myasthenia gravis. Nat. Clin. Pract. Neurol. 2008, 4, 317–327. [Google Scholar] [CrossRef] [PubMed]
  78. Higuchi, O.; Hamuro, J.; Motomura, M.; Yamanashi, Y. Autoantibodies to low density lipoprotein receptor-related protein 4 in myasthenia gravis. Ann. Neurol. 2011, 69, 418–422. [Google Scholar] [CrossRef]
  79. Grob, D.; Brunner, N.; Namba, T.; Pagala, M. Lifetime course of myasthenia gravis. Muscle Nerve 2008, 37, 141–149. [Google Scholar] [CrossRef] [PubMed]
  80. Kostera-Pruszczyk, A.; Kamińska, A.; Dutkiewicz, M.; Emeryk-Szajewska, B.; Strugalska-Cynowska, M.H.; Vincent, A.; Kwieciński, H. MuSK-positive myasthenia gravis is rare in the Polish population. Eur. J. Neurol. 2008, 15, 720–724. [Google Scholar] [CrossRef]
  81. Morren, J.; Li, Y. Maintenance immunosuppression in myasthenia gravis, an update. J. Neurol. Sci. 2020, 410, 116648. [Google Scholar] [CrossRef]
  82. Gold, R.; Hohlfeld, R.; Toyka, K.V. Progress in the treatment of myasthenia gravis. Ther. Adv. Neurol. Disord. 2008, 1, 99–114. [Google Scholar] [CrossRef] [Green Version]
  83. Sathasivam, S. Current and emerging treatments for the management of myasthenia gravis. Ther. Clin. Risk Manag. 2011, 7, 313–323. [Google Scholar] [CrossRef] [Green Version]
  84. Urban, P.P.; Jacobi, C.; Jander, S. Treatment Standards and Individualized Therapy of Myasthenia Gravis. Neurol. Int. Open 2018, 2, E84–E92. [Google Scholar] [CrossRef] [Green Version]
  85. Sieb, J.P. Myasthenia gravis: An update for the clinician. Clin. Exp. Immunol. 2014, 175, 408–418. [Google Scholar] [CrossRef] [PubMed]
  86. Hartmann, J.; Rivner, M.H. Methotrexate in myasthenia gravis. Clin. Neurophysiol. 2009, 120, e123–e124. [Google Scholar] [CrossRef]
  87. Gold, R.; Schneider-Gold, C. Current and future standards in treatment of myasthenia gravis. Neurotherapeutics 2008, 5, 535–541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Abdou, A.M. Methotrexate for treatment of myasthenia gravis. Neurology 2007, 62 (Suppl. S1), 300–301. [Google Scholar]
  89. Heckmann, J.M.; Rawoot, A.; Bateman, K.; Renison, R.; Badri, M. A single-blinded trial of methotrexate versus azathioprine as steroid-sparing agents in generalized myasthenia gravis. BMC Neurol. 2011, 11, 97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Pasnoor, M.; He, J.; Herbelin, L.; Burns, T.M.; Nations, S.; Bril, V.; Wang, A.K.; Elsheikh, B.H.; Kissel, J.T.; Saperstein, D.; et al. Methotrexate in MG Investigators of the Muscle Study Group. A randomized controlled trial of methotrexate for patients with generalized myasthenia gravis. Neurology 2016, 87, 57–64. [Google Scholar] [CrossRef] [Green Version]
  91. Smolen, J.S.; Aletaha, D.; McInnes, I.B. Rheumatoid arthritis. Lancet 2016, 388, 2023–2038. [Google Scholar] [CrossRef]
  92. Alamanos, Y.; Drosos, A.A. Epidemiology of adult rheumatoid arthritis. Autoimmun. Rev. 2005, 4, 130–136. [Google Scholar] [CrossRef]
  93. Kumar, P.; Yao, L.J.; Said, S.; Paleja, B.; van Loosdregt, J.; Chua, C.; Arkachaisri, T.; Consolaro, A.; Gattorno, M.; Martini, A.; et al. Molecular mechanisms of autophagic memory in pathogenic T cells in human arthritis. J. Autoimmun. 2018, 94, 90–98. [Google Scholar] [CrossRef]
  94. Mellado, M.; Martínez-Muñoz, L.; Cascio, G.; Lucas, P.; Pablos, J.L.; Rodríguez-Frade, J.M. T Cell Migration in Rheumatoid Arthritis. Front. Immunol. 2015, 6, 384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Lipsky, P.E.; van der Heijde, D.M.; St Clair, E.W.; Furst, D.E.; Breedveld, F.C.; Kalden, J.R.; Smolen, J.S.; Weisman, M.; Emery, P.; Feldmann, M.; et al. Infliximab and methotrexate in the treatment of rheumatoid arthritis. N. Engl. J. Med. 2000, 343, 1594–1602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Feldmann, M.; Brennan, F.M.; Foxwell, B.M.; Maini, R.N. The role of TNF alpha and IL-1 in rheumatoid arthritis. Curr. Dir. Autoimmun. 2001, 3, 188–199. [Google Scholar] [CrossRef]
  97. Farrugia, M.; Baron, B. The role of TNF-α in rheumatoid arthritis: A focus on regulatory T cells. J. Clin. Transl. Res. 2016, 2, 84–90. [Google Scholar] [CrossRef] [PubMed]
  98. Ponchel, F.; Burska, A.N.; Hunt, L.; Gul, H.; Rabin, T.; Parmar, R.; Buch, M.H.; Conaghan, P.G.; Emery, P. T-cell subset abnormalities predict progression along the Inflammatory Arthritis disease continuum: Implications for management. Sci. Rep. 2020, 10, 3669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Furst, D.E.; Koehnke, R.; Burmeister, L.F.; Kohler, J.; Cargill, I. Increasing methotrexate effect with increasing dose in the treatment of resistant rheumatoid arthritis. J. Rheumatol. 1989, 16, 313–320. [Google Scholar]
  100. Weinblatt, M.E. Methotrexate in rheumatoid arthritis: A quarter century of development. Trans. Am. Clin. Climatol. Assoc. 2013, 124, 16–25. [Google Scholar]
  101. Kaneko, Y.; Atsumi, T.; Tanaka, Y.; Inoo, M.; Kobayashi-Haraoka, H.; Amano, K.; Miyata, M.; Murakawa, Y.; Yasuoka, H.; Hirata, S.; et al. Comparison of adding tocilizumab to methotrexate with switching to tocilizumab in patients with rheumatoid arthritis with inadequate response to methotrexate: 52-week results from a prospective, randomised, controlled study (SURPRISE study). Ann. Rheum. Dis. 2016, 75, 1917–1923. [Google Scholar] [CrossRef]
  102. Hazlewood, G.S.; Barnabe, C.; Tomlinson, G.; Marshall, D.; Devoe, D.; Bombardier, C. Methotrexate monotherapy and methotrexate combination therapy with traditional and biologic disease modifying antirheumatic drugs for rheumatoid arthritis: Abridged Cochrane systematic review and network meta-analysis. BMJ 2016, 353, i1777. [Google Scholar] [CrossRef] [Green Version]
  103. McGeer, P.L.; Rogers, J.; McGeer, E.G.; Sibley, J. Anti-inflammatory drugs and Alzheimer disease. Lancet 1990, 335, 1037. [Google Scholar] [CrossRef]
  104. Judge, A.; Garriga, C.; Arden, N.K.; Lovestone, S.; Prieto-Alhambra, D.; Cooper, C.; Edwards, C.J. Protective effect of antirheumatic drugs on dementia in rheumatoid arthritis patients. Alzheimers Dement. 2017, 3, 612–621. [Google Scholar] [CrossRef] [PubMed]
  105. Zhou, M.; Xu, R.; Kaelber, D.C.; Gurney, M.E. Tumor Necrosis Factor (TNF) blocking agents are associated with lower risk for Alzheimer’s disease in patients with rheumatoid arthritis and psoriasis. PLoS ONE 2020, 15, e0229819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Chou, R.C.; Kane, M.; Ghimire, S.; Gautam, S.; Gui, J. Treatment for Rheumatoid Arthritis and Risk of Alzheimer’s Disease: A Nested Case-Control Analysis. CNS Drugs 2016, 30, 1111–1120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Haustein, U.F.; Rytter, M. Methotrexate in psoriasis: 26 years’ experience with low-dose long-term treatment. J. Eur. Acad. Dermatol. Venereol. 2000, 14, 382–388. [Google Scholar] [CrossRef] [PubMed]
  108. Shen, S.; O’Brien, T.; Yap, L.M.; Prince, H.M.; McCormack, C.J. The use of methotrexate in dermatology: A review. Australas. J. Dermatol. 2012, 53, 1–18. [Google Scholar] [CrossRef] [PubMed]
  109. Weinstein, G.D. Methotrexate. Ann. Intern. Med. 1977, 86, 199–204. [Google Scholar] [CrossRef]
  110. Busger op Vollenbroek, F.T.M.; Doggen, C.J.M.; Janssens, R.W.A.; Bernelot Moens, H.J. Dermatological guidelines for monitoring methotrexate treatment reduce drug-survival compared to rheumatological guidelines. PLoS ONE 2018, 13, e0194401. [Google Scholar] [CrossRef] [Green Version]
  111. Berthelot, C.; Rivera, A.; Duvic, M. Skin directed therapy for mycosis fungoides: A review. J. Drugs Dermatol. 2008, 7, 655–666. [Google Scholar]
  112. Kanwar, A.J.; De, D. Methotrexate for treatment of lichen planus: Old drug, new indication. J. Eur. Acad. Dermatol. Venereol. 2013, 27, e410–e413. [Google Scholar] [CrossRef]
  113. Puza, C.; Cardones, A.R. Concepts and controversies in the treatment of cutaneous lichen planus. G. Ital. Dermatol. Venereol. 2017, 152, 607–614. [Google Scholar] [CrossRef]
  114. Schutt, A.C.; Bullington, W.M.; Judson, M.A. Pharmacotherapy for pulmonary sarcoidosis: A Delphi consensus study. Respir. Med. 2010, 104, 717–723. [Google Scholar] [CrossRef] [Green Version]
  115. Baughman, R.P.; Culver, D.A.; Judson, M.A. A concise review of pulmonary sarcoidosis. Am. J. Respir. Crit. Care Med. 2011, 183, 573–581. [Google Scholar] [CrossRef] [Green Version]
  116. Goljan-Geremek, A.; Bednarek, M.; Franczuk, M.; Puścińska, E.; Nowiński, A.; Czystowska, M.; Kamiński, D.; Korzybski, D.; Stokłosa, A.; Kowalska, A.; et al. Methotrexate as a single agent for treating pulmonary sarcoidosis: A single centre real-life prospective study. Pneumonol. Alergol. Pol. 2014, 82, 518–533. [Google Scholar] [CrossRef] [PubMed]
  117. Feagan, B.G.; Rochon, J.; Fedorak, R.N.; Irvine, E.J.; Wild, G.; Sutherland, L.; Steinhart, A.H.; Greenberg, G.R.; Gillies, R.; Hopkins, M.; et al. Methotrexate for the treatment of Crohn’s disease. N. Engl. J. Med. 1995, 332, 292–297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Feagan, B.G.; Fedorak, R.N.; Irvine, E.J.; Wild, G.; Sutherland, L.; Steinhart, A.H.; Greenberg, G.R.; Koval, J.; Wong, C.J.; Hopkins, M.; et al. A comparison of methotrexate with placebo for the maintenance of remission in Crohn’s disease. N. Engl. J. Med. 2000, 342, 1627–1632. [Google Scholar] [CrossRef] [PubMed]
  119. Compston, A. The 150th anniversary of the first depiction of the lesions of multiple sclerosis. J. Neurol. Neurosurg. Psychiatry. 1988, 51, 1249–1252. [Google Scholar] [CrossRef] [Green Version]
  120. Compston, A.; Coles, A. Multiple sclerosis. Lancet 2008, 372, 1502–1517. [Google Scholar] [CrossRef]
  121. Calabresi, P.A.; Wilterdink, J.L.; Rogg, J.M.; Mills, P.; Webb, A.; Whartenby, K.A. An open-label trial of combination therapy with interferon beta-1a and oral methotrexate in MS. Neurology 2002, 58, 314–317. [Google Scholar] [CrossRef]
  122. Ashtari, F.; Savoj, M.R. Effects of low dose methotrexate on relapsing-remitting multiple sclerosis in comparison to Interferon β-1α: A randomized controlled trial. J. Res. Med. Sci. 2011, 16, 457–462. [Google Scholar]
  123. Stark, J.W.; Josephs, L.; Dulak, D.; Clague, M.; Sadiq, S.A. Safety of long-term intrathecal methotrexate in progersive forms of MS. Ther. Adv. Neurol. Disord. 2019, 12, 1–8. [Google Scholar] [CrossRef]
  124. Swerdlow, S.H. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissue, 4th ed.; International Agency for Research on Cancer: Lyon, France, 2008; (updated in 2016). [Google Scholar]
  125. Quintanilla-Martinez, L. The 2016 updated WHO classification of lymphoid neoplasias. Hematol. Oncol. 2017, 35 (Suppl. S1), 37–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Khan, Z.A.; Tripathi, R.; Mishra, B. Methotrexate: A detailed review on drug delivery and clinical aspects. Expert Opin. Drug Deliv. 2012, 9, 151–169. [Google Scholar] [CrossRef] [PubMed]
  127. Karami, F.; Ranjbar, S.; Ghasemi, Y.; Negahdaripour, M. Analytical methodologies for determination of methotrexate and its metabolites in pharmaceutical, biological and environmental samples. J. Pharm. 2019, 9, 373–391. [Google Scholar] [CrossRef] [PubMed]
  128. Mantadakis, E.; Cole, P.D.; Kamen, B.A. High-Dose Methotrexate in Acute Lymphoblastic Leukemia: Where Is the Evidence for Its Continued Use? Pharmacotherapy 2005, 25, 748–755. [Google Scholar] [CrossRef] [PubMed]
  129. Kamel, O.W. Lymphomas during Long-term Methotrexate Therapy. Arch. Dermatol. 1997, 133, 903–904. [Google Scholar] [CrossRef] [PubMed]
  130. Paul, C.; Le Tourneau, A.; Cayuela, J.M.; Devidas, A.; Robert, C.; Molinie, V.; Dubertret, L. Epstein-Barr Virus-Associated Lymphoproliferative Disease During Methotrexate Therapy for Psoriasis. Arch. Dermatol. 1997, 133, 867–871. [Google Scholar] [CrossRef]
  131. Alpdogan, O.; Kartan, S.; Johnson, W.; Sokol, K.; Porcu, P. Systemic therapy of cutaneous T-cell lymphoma (CTCL). Chin. Clin. Oncol. 2019, 8, 10. [Google Scholar] [CrossRef]
  132. Zackheim, H.S.; Kashani-Sabet, M.; McMillan, A. Low-dose methotrexate to treat mycosis fungoides: A retrospective study in 69 patients. J. Am. Acad. Dermatol. 2003, 49, 873–878. [Google Scholar] [CrossRef]
  133. Whittaker, S.; Hoppe, R.; Prince, H.M. How I treat mycosis fungoides and Sèzary syndrome. Blood 2016, 127, 3142–3153. [Google Scholar] [CrossRef] [Green Version]
  134. Olek-Hrab, K.; Maj, J.; Chmielowska, E.; Jankowska-Konsur, A.; Olszewska, B.; Kręcisz, B.; Iwankowski, P.; Mackiewicz-Wysocka, M.; Adamski, Z.; Nowicki, R.; et al. Methotrexate in the treatment of mycosis fungoides-a multicenter observational study in 79 patients. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 3586–3594. [Google Scholar] [CrossRef]
  135. Guo, Y.; Ahn, M.J.; Chan, A.; Wang, C.-H.; Kang, J.-H.; Kim, S.-B.; Bello, M.; Arora, R.S.; Zhang, Q.; He, X.; et al. Afatinib versus methotrexate as second-line treatment in Asian patients with recurrent or metastatic squamous cell carcinoma of the head and neck progressing on or after platinum-based therapy (LUX-Head & Neck 3): An open-label, randomised phase III trial. Ann. Oncol. 2019, 30, 1831–1839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Maiti, R. Metronomic chemotherapy. J. Pharmacol. Pharmacother. 2014, 5, 186–192. [Google Scholar] [CrossRef] [Green Version]
  137. Wen, J.; Ma, X.; Ruan, D.; Chen, J.; Lin, Q.; Dong, M.; Li, H.; Wu, H. The efficacy of metronomic thalimomide and methotrexate for refractory/relapsed non-Hodgkin lymphoma. J. Clin. Oncol. 2014, 32. [Google Scholar] [CrossRef]
  138. Bertino, J.B.; Mosher, M.B.; DeConti, R.C. Chemotherapy of cancer of the head and neck. Cancer 1973, 31, 1141–1149. [Google Scholar] [CrossRef]
  139. Chang, P.M.H.; Teng, H.W.; Chen, P.M.; Chang, S.Y.; Chu, P.Y.; Tsai, T.L.; Tai, S.K.; Wang, Y.F.; Huang, J.L.; Yang, M.H. Methotrexate and Leucovorin Double-Modulated 5-fluorouracil Combined With Cisplatin (MPFL) in Metastatic/Recurrent Head and Neck Cancer. J. Chin. Med. Assoc. 2008, 71, 336–341. [Google Scholar] [CrossRef] [Green Version]
  140. Kushwaha, V.S.; Gupta, S.; Husain, N.; Khan, H.; Negi, M.P.S.; Jamal, N.; Ghatak, A. Gefitinib, Methotrexate and Methotrexate plus 5-Fluorouracil as palliative treatment in recurrent head and neck squamous cell carcinoma. Cancer Biol. Ther. 2015, 16, 346–351. [Google Scholar] [CrossRef] [Green Version]
  141. Pai, P.S.; Vaidya, A.D.; Prabhash, K.; Banavali, S.D. Oral metronomic scheduling of anticancer therapy-based treatment compared to existing standard of care in locally advanced oral squamous cell cancers: A matched-pair analysis. Indian J. Cancer 2013, 50, 135–141. [Google Scholar] [CrossRef]
  142. Kumar, K.S.S.; Arumugam, V. Outcome of Oral Metronomic Therapy with Methotrexate and Celecoxib in Advanced/Recurrent Head and Neck Squamous Cell Carcinoma. Int. J. Sci. Study 2019, 6, 203–208. [Google Scholar]
  143. Mateen, A.; Adil, A.R.; Maken, R.N.; Khan, S.A.; Arif, M. Metronomic chemotherapy in recurrent head and neck cancer. J. Clin. Oncol. 2015, 33 (Suppl. abstr), e17007. [Google Scholar] [CrossRef]
  144. Denis, D.; Beneton, N.; Laribi, K.; Maillard, H. Management of mycosis fungoides-type cutaneous T-cell lymphoma (MF-CTCL): Focus on chlormethine gel. Cancer Manag. Res. 2019, 20, 2241–2251. [Google Scholar] [CrossRef] [Green Version]
  145. Aviles, A.; Neri, N.; Fernandez-Diez, J.; Silva, L.; Nambo, M. Interferon and low doses of methotrexate versus interferon and retinoids in the treatment of refractory/relapsed cutaneous T-cell lymphoma. Hematology 2015, 20, 538–542. [Google Scholar] [CrossRef] [PubMed]
  146. Wood, G.S.; Wu, J. Methotrexate and Pralatrexate. Dermatol. Clin. 2015, 33, 747–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Aviles, A.; Nambo, M.J.; Neri, N.; Castañeda, C.; Cleto, S.; Gonzalez, M.; Huerta-Guzmán, J. Interferon and low dose methotrexate improve outcome in refractory mycosis fungoides/Sézary syndrome. Cancer Biother. Radiopharm. 2007, 22, 836–840. [Google Scholar] [CrossRef] [PubMed]
  148. Kuo, Y.-H.; Chang, C.-H. Erythrodermic mycosis fungoides treated with low-dose methotrexate and 311 nm UV-B: A case report with 3-year follow up and literature review. Dermatol. Sin. 2016, 34, 37–41. [Google Scholar] [CrossRef] [Green Version]
  149. Raychaudhury, T. Management strategies for mycosis fungoides in India. Indian J. Dermatol. 2017, 62, 137–141. [Google Scholar] [CrossRef]
  150. Zakem, M.H.; Davis, B.R.; Adelstei, D.J.; Hines, J.D. Treatment of advanced stage mycosis fungoides with bleomycin, doxorubicin, and methotrexate with topical nitrogen mustard (BAM-M). Cancer 1986, 58, 2611–2616. [Google Scholar] [CrossRef]
  151. Vincent, R.G.; Wilson, H.E.; Lane, W.W.; Chen, T.Y.; Raza, S.; Gutierrez, A.C.; Caracandas, J.E. Progress in the Chemotherapy of Small Cell. Carcinoma of the Lung. Cancer 1981, 47, 229–235. [Google Scholar] [CrossRef]
  152. Hande, K.R.; Oldham, R.K.; Fer, M.F.; Richardson, R.L.; Greco, F.A. Randomized study of high-dose versus low-dose methotrexate in the treatment of extensive small cell lung cancer. Am. J. Med. 1982, 73, 413–419. [Google Scholar] [CrossRef]
  153. Neijstrom, E.S.; Capizzi, R.L.; Rudnick, S.A.; Kirsch, M.; Delaney, D.; Kahn, L.; Lipper, S.; Carney, C. High-dose methotrexate in small cell lung cancer. Lack of efficacy in preventing CNS relapse. Cancer 1983, 51, 1056–1061. [Google Scholar] [CrossRef]
  154. Preiss, R.; Brovtsyn, V.K.; Perevodchikova, N.L.; Bychkov, M.B.; Hüller, H.; Belova, L.A.; Michailov, P. Effect of methotrexate on the pharmacokinetics and renal excretion of cisplatin. Eur. J. Clin. Pharmacol. 1988, 34, 139–144. [Google Scholar] [CrossRef]
  155. Malzyner, A.; Caponero, R.; Donato, E.M.; Nadalin, W.; Vargas, F.S. Methotrexate (MTX), mitomycin C (MMC), vinorelbine (M) and cisplatinum (P) [M-MNP] in the treatment of advanced non-small cell lung cancer (NSCLC). Lung Cancer 1994, 11, 123. [Google Scholar] [CrossRef]
  156. Hall, R.R.; Bloom, H.J.G.; Freeman, J.E.; Nawrocki, A.; Wallace, D.M. Methotrexate Treatment for Advanced Bladder Cancer. Br. J. Urol. 1974, 46, 431–438. [Google Scholar] [CrossRef] [PubMed]
  157. Andrews, M.C.; Wilson, W.L. Phase II study of methotrexate (NSC-740) in solid tumors. Cancer Chemother. Rep. 1976, 51, 471–474. [Google Scholar]
  158. Turner, A.G.; Hendry, W.F.; Williams, G.B.; Bloom, H.J.G. The Treatment of Advanced Bladder Cancer with Methotrexate. Br. J. Urol. 1977, 49, 673–678. [Google Scholar] [CrossRef]
  159. Kaye, S.B.; McWhinnie, D.; Hart, A.; Deane, R.F.; Billaert, P.; Welsh, J.; Milsted, R.V.; Stuart, J.F.B.; Kalman, K.C. The treatment of advanced bladder cancer with methotrexate and cisplatinum—A pharmacokinetic study. Eur. J. Cancer Clin. Oncol. 1984, 20, 249–252. [Google Scholar] [CrossRef]
  160. Newling, D.W.W.; Stoter, G.; Sylvester, R.; de Pauw, M. The chemotherapy of advanced bladder cancer. Cancer Chemother. Pharmacol. 1987, 20, S39–S43. [Google Scholar] [CrossRef]
  161. Ismaili, N.; Amzerin, M.; Flechon, A. Chemotherapy in advanced bladder cancer: Current status and future. J. Hematol. Oncol. 2011, 4, 35. [Google Scholar] [CrossRef] [Green Version]
  162. Pavone-Macaluso, M.; Sternberg, C. Second-Line Chemotherapy in Advanced Bladder Cancer. Urol. Int. 2000, 64, 61–69. [Google Scholar] [CrossRef]
  163. Ahmed, T.; Yagoda, A.; Needles, B.; Scher, H.I.; Watson, R.C.; Geller, N. Vinblastine and methotrexate for advanced bladder cancer. J. Urol. 1985, 133, 602–604. [Google Scholar] [CrossRef]
  164. Sinha, A.K.; Anand, S.; Ortel, B.J.; Chang, Y.; Mai, Z.; Hasan, T.; Maytin, E.V. Methotrexate used in combination with aminolaevulinic acid for photodynamic killing of prostate cancer cells. Br. J. Cancer 2006, 95, 485–495. [Google Scholar] [CrossRef]
  165. Dimitriadis, I.; Bamias, A. Systemic Chemotherapy for Urothelial Cancer–How to Select Systemic Therapy in Bladder Cancer. Eur. Oncol. Haematol. 2017, 13, 134–138. [Google Scholar] [CrossRef] [Green Version]
  166. Sakura, T.; Hayakawa, F.; Sugiura, I.; Murayama, T.; Imai, K.; Usui, N.; Fujisawa, S.; Yamauchi, T.; Yujiri, T.; Kakihana, K.; et al. High-dose methotrexate therapy significantly improved survival of adult acute lymphoblastic leukemia: A phase III study by JALSG. Leukemia 2018, 32, 626–632. [Google Scholar] [CrossRef] [PubMed]
  167. van der Plas, E.; Nieman, B.J.; Butcher, D.T.; Hitzler, J.K.; Weksberg, R.; Ito, S.; Schachar, R. Neurocognitive late effects of chemotherapy in survivors of acute lymphoblastic leukemia: Focus on methotrexate. J. Can. Acad. Child. Adolesc. Psychiatry 2015, 24, 25–32. [Google Scholar] [PubMed]
  168. Hryniuk, W.M.; Bertino, J.R. Treatment of leukemia with large doses of methotrexate and folinic acid: Clinical-biochemical correlates. J. Clin. Investig. 1969, 48, 2140–2155. [Google Scholar] [CrossRef] [PubMed]
  169. Brenner, T.L.; Evans, W.E. Rationale for High-Dose Methotrexate in Childhood Acute Lymphoblastic Leukemia. In Treatment of Acute Leukemias, Antileukemic Drugs; Humana Press: Totowa, NJ, USA, 2003; pp. 339–356. [Google Scholar]
  170. Camitta, B.M.; Kamen, B.A. Role of Methotrexate in the Treatment of Acute Lymphoblastic Leukemia. In Treatment of Acute Leukemias: Antileukemic Drugs; Humana Press: Totowa, NJ, USA, 2003; pp. 357–362. [Google Scholar]
  171. Rizzoli, V.; Mangoni, L.; Caramatti, C.; Degliantoni, G.; Costi, D. High-dose methotrexate-leucovorin rescue therapy: Selected application in non-Hodgkin’s lymphoma. Tumori 1985, 71, 155–158. [Google Scholar] [CrossRef]
  172. Canellos, G.P.; Skarin, A.T.; Rosenthal, D.S.; Moloney, W.C.; Frei, E. Methotrexate as a single agent and in combination chemotherapy for the treatment of non-Hodgkin’s lymphoma of unfavorable histology. Cancer Treat. Rep. 1981, 65, 125–129. [Google Scholar]
  173. Khan, R.B.; Shi, W.; Thaler, H.T.; DeAngelis, L.M.; Abrey, L.E. Is Intrathecal Methotrexate Necessary in the Treatment of Primary CNS Lymphoma? J. Neurooncol. 2002, 58, 175–178. [Google Scholar] [CrossRef]
  174. Batchelor, T.T.; Kolak, G.; Ciordia, R.; Foster, C.S.; Henson, J.W. High-Dose Methotrexate for Intraocular Lymphoma. Clin. Cancer Res. 2003, 9, 711–715. [Google Scholar] [CrossRef]
  175. Fahey, J.B. High-Dose Methotrexate and Primary Central Nervous System Lymphoma. J. Neurosci. Nurs. 2007, 39, 83–88. [Google Scholar] [CrossRef]
  176. Zhu, J.-J.; Gerstner, E.R.; Engler, D.A.; Mrugala, M.M.; Nugent, W.; Nierenberg, K.; Hochberg, F.H.; Betensky, R.A.; Batchelor, T.T. High-dose methotrexate for elderly patients with primary CNS lymphoma. Neuro-Oncology 2009, 11, 211–215. [Google Scholar] [CrossRef] [Green Version]
  177. Gong, F.; Meng, Q.; Liu, C.; Zhao, Y. Efficacy and association analysis of high-dose methotrexate in the treatment of children with acute lymphoblastic leukemia. Oncol. Lett. 2019, 17, 4423–4428. [Google Scholar] [CrossRef] [PubMed]
  178. De Wilde, V.; Dierickx, D.; Schroyens, W.; Van Den Neste, E.; Bonnet, C.; Andre, M.; Janssens, A.; Van Hende, V.; Van Hoof, A. BHS guidelines for primary central nervous system lymphoma. Belg. J. Hematol. 2016, 7, 69–78. [Google Scholar]
  179. Bergner, N.; Monsef, I.; Illerhaus, G.; Engert, A.; Skoetz, N. Role of chemotherapy additional to high-dose methotrexate for primary central nervous system lymphoma (PCNSL). Cochrane Database Syst. Rev. 2012, 11. [Google Scholar] [CrossRef] [PubMed]
  180. Bokstein, F.; Lossos, A.; Lossos, I.S.; Siegal, T. Central Nervous System Relapse of Systemic Non-Hodgkin’s Lymphoma: Results of Treatment Based on High-dose Methotrexate Combination Chemotherapy. Leuk. Lymphoma 2002, 43, 587–593. [Google Scholar] [CrossRef] [PubMed]
  181. Omuro, A.M.; Taillandier, L.; Chinot, O.; Carnin, C.; Barrie, M.; Hoang-Xuan, K. Temozolomide and methotrexate for primary central nervous system lymphoma in the elderly. J. Neurooncol. 2007, 85, 207–211. [Google Scholar] [CrossRef]
  182. Sun, X.; Liu, J.; Wang, Y.; Bai, X.; Chen, Y.; Qian, J.; Zhu, H.; Liu, F.; Qiu, X.; Sun, S.; et al. Methotrexate-cytarabine-dexamethasone combination chemotherapy with or without rituximab in patients with primary central nervous system lymphoma. Oncotarget 2017, 8, 49156–49164. [Google Scholar] [CrossRef]
  183. Reinius, M.; Ahmad, S.S.; Crawley, C.; Williams, M.V.; Wimperis, J.; Follows, G.A. High-Dose Methotrexate in the Treatment of Primary Testicular Lymphoma. Blood 2013, 122, 5108. [Google Scholar] [CrossRef]
  184. Morris, P.G.; Correa, D.D.; Yahalom, J.; Raizer, J.J.; Schiff, D.; Grant, B.; Grimm, S.; Lai, R.K.; Reiner, A.S.; Panageas, K.; et al. Rituximab, Methotrexate, Procarbazine, and Vincristine Followed by Consolidation Reduced-Dose Whole-Brain Radiotherapy and Cytarabine in Newly Diagnosed Primary CNS Lymphoma: Final Results and Long-Term Outcome. J. Clin. Oncol. 2013, 31, 3971–3979. [Google Scholar] [CrossRef]
  185. Kansara, R.R.; Shenkier, T.; Connors, J.M.; Gerrie, A.S.; Klasa, R.; Savage, K.J.; Sehn, L.H.; Villa, D. Rituximab with High Dose Methotrexate in the Management of Primary Central Nervous System Diffuse Large B-Cell Lymphoma. Blood 2014, 124, 3090. [Google Scholar] [CrossRef]
  186. Inaly, K.; Crew, L.L.; Graham, C.A.; Mrugala, M.M. Primary central nervous system lymphoma treated with high-dose methotrexate and rituximab: A single-institution experience. Oncol. Lett. 2016, 11, 3471–3476. [Google Scholar] [CrossRef] [Green Version]
  187. BC Cancer Protocol Summary LYHDMRP. BC Cancer Protocol Summary for Treatment of Primary Intracerebral Lymphoma with High Dose Methotrexate and rituximab. Available online: http://www.bccancer.bc.ca/chemotherapy-protocols-site/Documents/Lymphoma-Myeloma/LYHDMRP_Protocol.pdf (accessed on 2 January 2020).
  188. Buechler, M.; Mukherji, B.; Chasin, W.; Nathanson, L. High dose methotrexate with and without BCG therapy in advanced head and neck malignancy. Cancer 1979, 43, 1095–1100. [Google Scholar] [CrossRef]
  189. Schappell, D.L.; Alper, J.C.; McDonald, C.J. Treatment of Advanced Mycosis Fungoides and Sézary Syndrome with Continuous Infusions of Methotrexate Followed by Fluorouracil and Leucovorin Rescue. Arch. Dermatol. 1995, 131, 307–313. [Google Scholar] [CrossRef] [PubMed]
  190. Singh, V.K.; Subudhi, B.B. Development of reversible glutamine conjugate of methotrexate for enhanced brain delivery. Med. Chem. Res. 2015, 24, 624–635. [Google Scholar] [CrossRef]
  191. Singh, V.K.; Subudhi, B.B. Development and characterization of lysine-methotrexate conjugate for enhanced brain delivery. Drug Deliv. 2016, 23, 2327–2337. [Google Scholar] [CrossRef]
  192. Corem-Salkmon, E.; Ram, Z.; Daniels, D.; Perlstein, B.; Last, D.; Salomon, S.; Tamar, G.; Shneor, R.; Guez, D.; Margel, S.; et al. Convection-enhanced delivery of methotrexate-loaded maghemite nanoparticles. Int. J. Nanomed. 2011, 6, 1595–1602. [Google Scholar] [CrossRef] [Green Version]
  193. Gagliardi, M.; Bardi, G.; Bifone, A. Polymeric nanocarriers for controlled and enhanced delivery of therapeutic agents to the CNS. Ther. Deliv. 2012, 3, 875–887. [Google Scholar] [CrossRef]
  194. Wosikowski, K.; Biedermann, E.; Rattel, B.; Breiter, N.; Jank, P.; Löser, R.; Jansen, G.; Peters, G.J. Vivo Antitumor Activity of Methotrexate Conjugated to Human Serum Albumin in Human Cancer Cells. Clin. Cancer Res. 2003, 9, 1917–1926. [Google Scholar]
  195. Burger, A.M.; Hartung, G.; Stehle, G.; Sinn, H.; Fiebig, H.H. Pre-clinical Evaluation of a Methotrexate-Albumin Conjugate (MTX-HSA) in Human Tumor Xenografts in Vivo. Int. J. Cancer 2001, 92, 718–724. [Google Scholar] [CrossRef]
  196. Kohler, N.; Sun, C.; Wang, J.; Zhang, M. Methotrexate-Modified Superparamagnetic Nanoparticles and Their Intracellular Uptake into Human Cancer Cells. Langmuir 2005, 21, 8858–8864. [Google Scholar] [CrossRef]
  197. Gao, K.; Jiang, X. Influence of particle size on transport of methotrexate across blood brain barrier by polysorbate 80-coated polybutylcyanoacrylate nanoparticles. Int. J. Pharm. 2006, 310, 213–219. [Google Scholar] [CrossRef]
  198. Trapani, A.; Denora, N.; Iacobellis, G.; Sitterberg, J.; Bakowsky, U.; Kissel, T. Methotrexate-loaded chitosan- and glycol chitosan-based nanoparticles: A promising strategy for the administration of the anticancer drug to brain tumors. AAPS Pharm. Sci. Tech. 2011, 12, 1302–1311. [Google Scholar] [CrossRef] [Green Version]
  199. Mc Carthy, D.J.; Malhotra, M.; O’Mahony, A.M.; Cryan, J.F.; O’Driscoll, C.M. Nanoparticles and the Blood-Brain Barrier: Advancing from In-Vitro Models towards Therapeutic Significance. Pharm. Res. 2015, 32, 1161–1185. [Google Scholar] [CrossRef] [PubMed]
  200. Kosasih, A.; Bowman, B.J.; Wigent, R.J.; Ofner, C.M., III. Characterization and in Vitro Release of Methotrexate From Gelatin/Methotrexate Conjugates Formed Using Different Preparation Variables. Int. J. Pharm. 2000, 204, 81–89. [Google Scholar] [CrossRef]
  201. Narayani, R.; Rao, K.P. Controlled release of anticancer drug methotrexate from biodegradable gelatin microspheres. J. Microencapsul. 1994, 11, 69–77. [Google Scholar] [CrossRef] [PubMed]
  202. Narayani, R.; Rao, K.P. Biodegradable microspheres using two different gelatin drug conjugates for the controlled delivery of methotrexate. Int. J. Pharm. 1996, 128, 261–268. [Google Scholar] [CrossRef]
  203. Pica, K.; Tchao, R.; Ofner, C.M., III. Gelatin–methotrexate conjugate microspheres as a potential drug delivery system. J. Pharm. Sci. 2006, 95, 1896–1908. [Google Scholar] [CrossRef]
  204. Ofner, C.M., III; Pica, K.; Bowman, B.J.; Chen, C.S. Growth inhibition, drug load, and degradation studies of gelatin/methotrexate conjugates. Int. J. Pharm. 2006, 308, 90–99. [Google Scholar] [CrossRef]
  205. Nagy, G.; Király, G.; Veres, P.; Lázár, I.; Fábián, I.; Bánfalvi, G.; Juhász, I.; Kalmár, J. Controlled release of methotrexate from functionalized silica-gelatin aerogel microparticles applied against tumor cell growth. Int. J. Pharm. 2019, 558, 396–403. [Google Scholar] [CrossRef]
  206. Kukowska-Latallo, J.F.; Candido, K.A.; Cao, Z.; Nigavekar, S.S.; Majoros, I.J.; Thomas, T.P.; Balogh, L.P.; Khan, M.K.; Baker, J.R., Jr. Nanoparticle Targeting of Anticancer Drug Improves Therapeutic Response in Animal Model of Human Epithelial Cancer. Cancer Res. 2005, 65, 5317–5324. [Google Scholar] [CrossRef] [Green Version]
  207. Majoros, I.J.; Myc, A.; Thomas, T.; Mehta, C.B.; Baker, J.R., Jr. PAMAM dendrimer-based multifunctional conjugate for cancer therapy: Synthesis, characterization, and functionality. Biomacromolecules 2006, 7, 572–579. [Google Scholar] [CrossRef]
  208. Wu, G.; Barth, R.F.; Yang, W.; Kawabata, S.; Zhang, L.; Green-Church, K. Targeted delivery of methotrexate to epidermal growth factor receptor–positive brain tumors by means of cetuximab (IMC-C225) dendrimer bioconjugates. Mol. Cancer Ther. 2006, 5, 52–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Bhattacharya, S.; Alkharfy, K.M.; Janardhanan, R.; Mukhopadhyay, D. Nanomedicine: Pharmacological perspectives. Nanotechnol. Rev. 2012, 1, 235–253. [Google Scholar] [CrossRef]
  210. Dhanikula, R.S.; Hildgen, P. Influence of molecular architecture of polyether-co-polyester dendrimers on the encapsulation and release of methotrexate. Biomaterials 2007, 28, 3140–3152. [Google Scholar] [CrossRef]
  211. Riebeseel, K.; Biedermann, E.; Löser, R.; Breiter, N.; Hanselmann, R.; Mülhaupt, R.; Unger, C.; Kratz, F. Polyethylene glycol conjugates of methotrexate varying in their molecular weight from MW 750 to MW 40000: Synthesis, characterization, and structure-activity relationships in vitro and in vivo. Bioconjug. Chem. 2002, 13, 773–785. [Google Scholar] [CrossRef] [PubMed]
  212. Kmiecik, S.; Krzyścik, M.; Filip-Psurska, B.; Wietrzyk, J.; Boratyński, J.; Goszczyński, T.M. Methotrexate and epirubicin conjugates as potential antitumor drugs. Adv. Clin. Exp. Med. 2017, 71, 618–623. [Google Scholar] [CrossRef]
  213. Martin, M.; Casado, A.; López-Martin, J.A.; Rodriguez-lescure, A.; Nieto, Y.; Ayala, F.; Diaz-rubio, E. Preliminary results. UFT/Methotrexate/Leucovorin for Breast Cancer Patients in Progression after HDCT/PBPC Support. Oncology (Williston Park) 1997, 11, 83–85. [Google Scholar]
  214. Martin, M.; Casado, A.; Macias, J.A.; Garcia-Carbonero, I.; Garcia-Saenz, J.A.; Oruezabal, M.; Manrique, I.; Diaz-Rubio, E. Methotrexate, uracil and tegafur, and leucovorin chemotherapy for patients with breast cancer in progression after high-dose chemotherapy with peripheral blood progenitor cell transplant: A phase II study. Am. J. Clin. Oncol. 2000, 23, 617–621. [Google Scholar] [CrossRef]
  215. Moreno, A.; Moreno, A.; Escobedo, A.; Benito, E.; Serra, J.M.; Guma, A.; Riu, F. Pathologic Changes Related to CMF Primary Chemotherapy in Breast Cancer. Breast Cancer Res. Treat. 2002, 75, 119–125. [Google Scholar] [CrossRef]
  216. Cocconi, G.; Blasio, B.D.; Boni, C.; Bisagni, G.; Rondini, E.; Bella, M.A.; Leonardi, F.; Savoldi, L.; Vallisneri, C.; Camisa, R.; et al. Primary chemotherapy in operable breast carcinoma comparing CMF (cyclophosphamide, methotrexate, 5-fluorouracil) with an anthracycline-containing regimen: Short-term responses translated into long-term outcomes. Ann. Oncol. 2005, 16, 1469–1476. [Google Scholar] [CrossRef]
  217. Leone, J.P.; Leone, J.; Vallejo, C.T.; Pérez, J.E.; Romero, A.O.; Machiavelli, M.R.; Acuña, L.R.; Domínguez, M.E.; Langui, M.; Fasce, H.M.; et al. Sixteen years follow-up results of a randomized phase II trial of neoadjuvant fluorouracil, doxorubicin, and cyclophosphamide (FAC) compared with cyclophosphamide, methotrexate, and 5-fluorouracil (CMF) in stage III breast cancer: GOCS experience. Breast Cancer Res. Treat. 2014, 143, 313–332. [Google Scholar] [CrossRef]
  218. Colleoni, M.; Rocca, A.; Sandri, M.T.; Zorzino, L.; Masci, G.; Nolè, F.; Peruzzotti, G.; Robertson, C.; Orlando, L.; Cinieri, S.; et al. Low-dose oral methotrexate and cyclophosphamide in metastatic breast cancer: Antitumor activity and correlation with vascular endothelial growth factor levels. Ann. Oncol. 2002, 13, 73–80. [Google Scholar] [CrossRef] [PubMed]
  219. Fukuda, T.; Tanabe, M.; Kobayashi, K.; Fukada, I.; Takahashi, S.; Iwase, T.; Ito, Y. Combination chemotherapy with mitomycin C and methotrexate is active against metastatic HER2-negative breast cancer even after treatment with anthracycline, taxane, capecitabine, and vinorelbine. Springerplus 2015, 4, 376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Cheng, X.I.; Zhou, T.Y.; Li, B.; Li, M.; Li, L.; Li, Z.; Lu, W. Methotrexate and 5-aminoimidazole-4-carboxamide riboside exert synergistic anticancer action against human breast cancer and hepatocellular carcinoma. Acta Pharmacol. Sin. 2013, 34, 951–959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Wu, C.W.; Liu, H.C.; Yu, Y.L.; Hung, Y.T.; Wei, C.W.; Yiang, G.T. Combined treatment with vitamin C and methotrexate inhibits triple-negative breast cancer cell growth by increasing H2O2 accumulation and activating caspase-3 and p38 pathways. Oncol. Rep. 2017, 37, 2177–2184. [Google Scholar] [CrossRef] [Green Version]
  222. Bhasin, R.R.; Nogeirie, C.P. Combination Chemotherapy (High-Dose Methotrexate with Citrovorum Factor Rescue, Mechlorethamine, and Procarbazine) in Non-Small Cell Lung Cancer. Cancer Chemother. Pharmacol. 1983, 11, 106–107. [Google Scholar] [CrossRef]
  223. Chen, J.; Yang, X.; Huang, L.; Lai, H.; Gan, C.; Luo, X. Development of dual-drug-loaded stealth nanocarriers for targeted and synergistic anti-lung cancer efficacy. Drug Deliv. 2018, 25, 1932–1942. [Google Scholar] [CrossRef]
  224. Kern, S.; Truebenbach, I.; Höhn, M.; Gorges, J.; Kazmaier, U.; Zahler, S.; Vollmar, A.M.; Wagner, E. Combined antitumoral effects of pretubulysin and methotrexate. Pharmacol. Res. Perspect. 2019, 7, e00460. [Google Scholar] [CrossRef]
  225. Yan, K.; Lee, L.; Hsieh, M.; Yan, M.; Yao, C.; Chang, P.; Chen, T.; Chang, H.; Cheng, A.; Lai, G.; et al. Aspirin antagonizes the cytotoxic effect of methotrexate in lung cancer cells. Oncol. Rep. 2013, 30, 1497–1505. [Google Scholar] [CrossRef]
  226. Abdelrady, H.; Hathout, R.M.; Osman, R.; Saleem, I.; Mortada, N.D. Exploiting gelatin nanocarriers in the pulmonary delivery of methotrexate for lung cancer therapy. Eur. J. Pharm. Sci. 2019, 133, 115–126. [Google Scholar] [CrossRef]
  227. Kubota, Y.; Shuin, T.; Miura, T.; Moriyama, M.; Fukushima, S.; Shiozaki, H. Vincristine, bleomycin, cyclophosphamide, mitomycin, and methotrexate. Cancer Treat. Rep. 1984, 68, 1167–1168. [Google Scholar]
  228. Hall, R.R.; Newling, D.W.W.; Ramsden, P.D.; Richards, B.; Robinson, M.R.G.; Phillips, P.A. Treatment of invasive bladder cancer by local resection and high dose Methotrexate. Br. J. Urol. 1984, 56, 668–672. [Google Scholar] [CrossRef] [PubMed]
  229. Jaffe, N.; Gorlick, R. High-Dose Methotrexate in Osteosarcoma: Let the Questions Surcease—Time for Final Acceptance. J. Clin. Oncol. 2008, 26, 4365–4366. [Google Scholar] [CrossRef] [PubMed]
  230. Bielack, S.S.; Smeland, S.; Whelan, J.S.; Marina, N.M.; Jovic, G.; Hook, J.M.; Krailo, M.; Gebhardt, M.C.; Papai, Z.; Mayer, J.; et al. Methotrexate, Doxorubicin, and Cisplatin (MAP) Plus Maintenance Pegylated Interferon Alfa-2b Versus MAP Alone in Patients With Resectable High-Grade Osteosarcoma and Good Histologic Response to Preoperative MAP: First Results of the EURAMOS-1 Good Response Randomized Controlled Trial. J. Clin. Oncol. 2015, 33, 2279–2287. [Google Scholar] [CrossRef] [PubMed]
  231. Krailo, M.; Ertel, I.; Makley, J.; Fryjer, C.J.; Baum, E.; Weetman, R.; Yunis, E.; Barnes, L.; Bleyer, W.A.; Hammond, G.D. A randomized study comparing high-dose methotrexate with moderate-dose methotrexate as components of adjuvant chemotherapy in childhood nonmetastatic osteosarcoma: A report from the Childrens Cancer Study Group. Med. Pediatr. Oncol. 1987, 15, 69–77. [Google Scholar] [CrossRef]
  232. Meyers, P.A.; Schwartz, C.L.; Krailo, M.; Kleinerman, E.S.; Betcher, D.; Bernstein, M.L.; Conrad, E.; Ferguson, W.; Gebhardt, M.; Goorin, A.M.; et al. Osteosarcoma: A randomized, prospective trial of the addition of ifosfamide and/or muramyl tripeptide to cisplatin, doxorubicin, and high-dose methotrexate. J. Clin. Oncol. 2005, 23, 2004–2011. [Google Scholar] [CrossRef] [PubMed]
  233. Winkler, K.; Beron, G.; Delling, G.; Heise, U.; Kabisch, H.; Purfurst, C.; Berger, J.; Ritter, J.; Jurgens, H.; Gerein, V. Neoadjuvant chemotherapy of osteosarcoma: Results of a randomized cooperative trial (COSS-82) with salvage chemotherapy based on histological tumor response. J. Clin. Oncol. 1988, 6, 329–337. [Google Scholar] [CrossRef]
  234. Bacci, G.; Picci, P.; Ferrari, S.; Ruggieri, P.; Casadei, R.; Tienghi, A.; Brach del Prever, A.; Gherlinzoni, F.; Mercuri, M.; Monti, C. Primary chemotherapy and delayed surgery for nonmetastatic osteosarcoma of the extremities. Results in 164 patients preoperatively treated with high doses of methotrexate followed by cisplatin and doxorubicin. Cancer 1993, 72, 3227–3238. [Google Scholar] [CrossRef]
  235. Bacci, G.; Ferrari, S.; Bertoni, F.; Ruggieri, P.; Picci, P.; Longhi, A.; Casadei, R.; Fabbri, N.; Forni, C.; Versari, M.; et al. Long-term outcome for patients with nonmetastatic osteosarcoma of the extremity treated at the istituto ortopedico rizzoli according to the istituto ortopedico rizzoli/osteosarcoma-2 protocol: An updated report. J. Clin. Oncol. 2000, 18, 4016–4027. [Google Scholar] [CrossRef]
  236. Geller, D.S.; Richard Gorlick, R. Osteosarcoma: A Review of Diagnosis, Management, and Treatment Strategies. Clin. Adv. Hematol. Oncol. 2010, 8, 705–718. [Google Scholar]
  237. Abrão, R.; de Andrade, J.; Tiezzi, D.; Marana, H.; dos Reis, F.C.; Clagnan, W. Treatment for Low-Risk Gestational Trophoblastic Disease: Comparison of Single-Agent Methotrexate, Dactinomycin and Combination Regimens. Gynecol. Oncol. 2008, 108, 149–153. [Google Scholar] [CrossRef]
  238. Han, S.N.; Amant, F.; Leunen, K.; Devi, U.K.; Neven, P.; Berteloot, P.; Vergote, I. Treatment of high-risk gestational trophoblastic neoplasia with weekly high-dose methotrexate-etoposide. Gynecol. Oncol. 2012, 127, 47–50. [Google Scholar] [CrossRef]
  239. Nebraska Lymphoma Study Group. Treatment of Non-Hodgkin’s Lymphoma and Hodgkin’s Lymphoma. University of Nebraska Medical Center, Department of Internal Medicine. 2009. Available online: https://www.unmc.edu/media/lymphoma/ne_lymphoma_book.pdf (accessed on 2 January 2020).
  240. Malcolm, T.I.M.; Hodson, D.J.; Macintyre, E.A.; Turner, S.D. Challenging perspectives on the cellular origins of lymphoma. Open Biol. 2016, 6, 160232. [Google Scholar] [CrossRef] [Green Version]
  241. Burkhardt, B. Paediatric lymphoblastic T-cell leukaemia and lymphoma: One or two diseases? Br. J. Haematol. 2009, 149, 653–668. [Google Scholar] [CrossRef] [PubMed]
  242. Davis, T.P.; Enna, S.J.; Lecount, L. Pharmacology of the Blood Brain Barrier: Targeting CNS Disorders, 1st ed.; Elsevier INC: Amsterdam, The Netherlands, 2014. [Google Scholar]
  243. Cho, K.M.; Kim, Y.J.; Kim, S.H.; Kim, J.W.; Lee, J.O.; Han, J.H.; Lee, K.W.; Kim, J.H.; Kim, C.Y.; Bang, S.M.; et al. Salvage Treatment with Intracerebrospinal Fluid Thiotepa in Patients with Leptomeningeal Metastasis After Failure of Methotrexate-based Treatment. Anticancer Res. 2015, 35, 5631–5638. [Google Scholar] [PubMed]
  244. Kapke, J.T.; Schneidewend, R.J.; Jawa, Z.A.; Chiang-Ching, H.; Connelly, J.M.; Chitambar, C.R. High-dose intravenous methotrexate in the management of breast cancer with leptomeningeal disease: Case series and review of the literature. Hematol. Oncol. Stem Cell Ther. 2019, 12, 189–193. [Google Scholar] [CrossRef]
  245. Kasim, A.; Whitehouse, M.; Ramachandran, C.; Sanz, M.B.; Lennernäs, H.; Hussain, A.S.; Junginger, H.E.; Stavchansky, S.; Midha, K.K.; Shah, V.P.; et al. Molecular Properties of WHO Essential Drugs and Provisional Biopharmaceutical Classification. Mol. Pharm. 2004, 1, 85–96. [Google Scholar] [CrossRef] [PubMed]
  246. Torchilin, V.P. Multifunctional Pharmaceutical Nanocarriers, 1st ed.; Springer: Boston, MA, USA, 2008. [Google Scholar]
  247. Martin-Banderas, L.; Holgado, M.A.; Venero, J.L.; Alvarez-Fuentes, J.; Fernández-Arévalo, M. Nanostructures for Drug Delivery to the Brain. Curr. Med. Chem. 2011, 18, 5303–5321. [Google Scholar] [CrossRef]
  248. Ricci, M.; Blasi, P.; Giovagnoli, S.; Rossi, C. Delivering drugs to the central nervous system: A medicinal chemistry or a pharmaceutical technology issue? Curr. Med. Chem. 2006, 13, 1757–1775. [Google Scholar] [CrossRef]
  249. Jang, J.H.; Jeong, S.H.; Lee, Y.B. Preparation and In Vitro/In Vivo Characterization of Polymeric Nanoparticles Containing Methotrexate to Improve Lymphatic Delivery. Int. J. Mol. Sci. 2019, 20, 3312. [Google Scholar] [CrossRef] [Green Version]
  250. Leone, L.A.; Albala, M.M.; Rege, V.B. Treatment of carcinoma of head and neck with intravenous methotrexate. Cancer 1968, 21, 828–837. [Google Scholar] [CrossRef]
  251. Capizzi, R.L.; DeConti, R.C.; Marsh, J.C.; Bertino, J.R. Methotrexate Therapy of Head and Neck Cancer: Improvement in Therapeutic Index by the Use of Leucovorin “Rescue”. Cancer Res. 1970, 30, 1782–1788. [Google Scholar] [PubMed]
  252. Pant, G.C.; Sanyal, B.; Khanna, N.N.; Rastogi, B.L. Sequential treatment of advanced oral carcinoma by methotrexate and radiotherapy. Indian J. Med. Res. 1973, 61, 848–855. [Google Scholar] [PubMed]
  253. Frei, E., III; Blum, R.H.; Pitman, S.W.; Kirkwood, J.M.; Henderson, I.C.; Skarin, A.T.; Mayer, R.S.; Bast, R.C.; Garnick, M.B.; Parker, L.M.; et al. High dose methotrexate with leucovorin rescue. Rationale and spectrum of antitumor activity. Am. J. Med. 1980, 68, 370–376. [Google Scholar] [CrossRef]
  254. Woods, R.L.; Fox, R.M.; Tattersall, M.H. Methotrexate treatment of squamous-cell head and neck cancers: Dose-response evaluation. Br. Med. J. (Clin. Res. Ed). 1981, 282, 600–602. [Google Scholar] [CrossRef] [Green Version]
  255. DeConti, R.C.; Schoenfeld, D. A randomized prospective comparison of intermittent methotrexate, methotrexate with leucovorin, and a methotrexate combination in head and neck cancer. Cancer 1981, 48, 1061–1072. [Google Scholar] [CrossRef]
  256. Hong, W.K.; Schaefer, S.; Issell, B.; Cummings, C.; Luedke, D.; Bromer, R.; Fotonoff, S.D.; Aoust, J.; Shapsay, S.; Welch, J.; et al. A prospective randomized trial of methotrexate versus cisplatin in the treatment of recurrent squamous cell carcinoma of the head and neck. Cancer 1983, 52, 206–210. [Google Scholar] [CrossRef]
  257. Drelichman, A.; Cummings, G.; Al-Sarraf, M. A randomized trial of the combination of cis-platinum, oncovin and bleomycin (COB) versus methotrexate in patients with advanced squamous cell carcinoma of the head and neck. Cancer 1983, 52, 399–403. [Google Scholar] [CrossRef]
  258. Pradhan, S.; Asthana, A.K.; Jain, R.K.; Pant, G.C.; Sahni, K. Single agent methotrexate chemotherapy for head and neck cancers. Indian J. Otolaryngol 1991, 43, 175. [Google Scholar] [CrossRef]
  259. Vermorken, J.B.; Specenier, P. Optimal treatment for recurrent/metastatic head and neck cancer. Ann. Oncol. 2010, 21, 252–261. [Google Scholar] [CrossRef]
  260. Pathak, A.B.; Kulkarni, H. Weekly methotrexate in squamous cell cancers of head and neck. J. Clin. Oncol. 2006, 24, 15539. [Google Scholar] [CrossRef]
  261. Banipal, R.P.S.; Mahajan, M.K. Methotrexate Revisited—in Recurrent Head and Neck Cancer. Palliat. Care Res. Treat. 2011, 5, 9–13. [Google Scholar] [CrossRef]
  262. Sharma, M.; Gupta, M.; Fotedar, V.; Sharma, A. Methotrexate, an attractive agent for palliation in head and neck cancers. South Asian J. Cancer 2014, 3, 229. [Google Scholar] [CrossRef] [PubMed]
  263. Gasparini, G. Metronomic scheduling: The future of chemotherapy? Lancet Oncol. 2001, 2, 733–740. [Google Scholar] [CrossRef]
  264. De Felice, F.; Musio, D.; Tombolini, V. Head and neck cancer: Metronomic chemotherapy. BMC Cancer 2015, 15, 677. [Google Scholar] [CrossRef] [Green Version]
  265. Schwartz, M.A. High-dose methotrexate for metastatic breast cancer to the central nervous system: A single-institution review of 46 patients. J. Clin. Oncol. 2014, 32, 1079. [Google Scholar] [CrossRef]
  266. Colleoni, M.; Cole, B.F.; Viale, G.; Regan, M.M.; Price, K.N.; Maiorano, E.; Mastropasqua, M.G.; Crivellari, D.; Gelber, R.D.; Goldhirsch, A.; et al. Classical cyclophosphamide, methotrexate, and fluorouracil chemotherapy is more effective in triple-negative, node-negative breast cancer: Results from two randomized trials of adjuvant chemoendocrine therapy for nodenegative breast cancer. J. Clin. Oncol. 2010, 28, 2966–2973. [Google Scholar] [CrossRef] [Green Version]
  267. Wu, C.E.; Chen, S.C.; Lin, Y.C.; Lo, Y.F.; Hsueh, S.; Chang, H.K. Identification of patients with node-negative, triple-negative breast cancer who benefit from adjuvant cyclophosphamide, methotrexate, and 5-fluorouracil chemotherapy. Anticancer. Res. 2014, 34, 1301–1306. [Google Scholar]
  268. Kumaki, N.; Okamatsu, C.; Tokuda, Y.; Nakamura, N. Breast Cancer in Patients of Rheumatoid Arthritis with Methotrexate Therapy Mimicking Histopathological Changes after Neoadjuvant Chemotherapy. Tokai J. Exp. Clin. Med. 2017, 42, 104–108. [Google Scholar]
  269. Hoeng, J.; Maeder, S.; Vanscheeuwijck, P.; Peitsch, M.C. Assessing the lung cancer risk reduction potential of candidate modified risk tobacco products. Intern. Emerg. Med. 2019, 14, 821–834. [Google Scholar] [CrossRef] [Green Version]
  270. Tomoda, R.; Seto, M.; Hioki, Y.; Sonoda, J.; Matsumine, A.; Kusuzaki, K.; Uchida, A. Low-dose Methotrexate Inhibits Lung Metastasis and Lengthens Survival in Rat Osteosarcoma. Clin. Exp. Metastasis 2005, 22, 559–564. [Google Scholar] [CrossRef]
  271. Sekimura, A.; Iwai, S.; Funasaki, A.; Motono, N.; Usuda, K.; Uramoto, H. Lung cancer combined with methotrexate-associated lymphoproliferative disorder: A case report. Int. J. Surg. Case Rep. 2019, 59, 161–164. [Google Scholar] [CrossRef] [PubMed]
  272. deKernion, J.B. The Chemotherapy of Advanced Bladder Carcinoma. Cancer Res. 1977, 37, 2771–2774. [Google Scholar] [PubMed]
  273. Osasan, S.; Zhang, M.; Shen, F.; Paul, P.J.; Persad, S.; Sergi, C. Osteogenic Sarcoma: A 21st Century Review. Anticancer Res. 2016, 36, 4391–4398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Xu, M.; Xu, S.F.; Yu, X.C. Clinical analysis of osteosarcoma patients treated with high-dose methotrexate-free neoadjuvant chemotherapy. Curr. Oncol. 2014, 21, e678–e684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Wippel, B.; Gundle, K.R.; Dang, T.; Paxton, J.; Bubalo, J.; Stork, L.; Fu, R.; Ryan, C.W.; Davis, L.E. Safety and Efficacy of High-Dose Methotrexate for Osteosarcoma in Adolescents Compared With Young Adults. Cancer Med. 2019, 8, 111–116. [Google Scholar] [CrossRef] [Green Version]
  276. Guo, W.; Healey, J.H.; Meyers, P.A.; Ladanyi, M.; Huvos, A.G.; Bertino, J.R.; Gorlick, R. Mechanisms of methotrexate resistance in osteosarcoma. Clin. Cancer. Res. 1999, 5, 621–627. [Google Scholar]
  277. Wang, J.; Li, G. Mechanisms of methotrexate resistance in osteosarcoma cell lines and strategies for overcoming this resistance. Oncol. Lett. 2014, 9, 940–944. [Google Scholar] [CrossRef] [Green Version]
  278. Xu, W.; Li, Z.; Zhu, X.; Xu, R.; Xu, Y. miR-29 Family Inhibits Resistance to Methotrexate and Promotes Cell Apoptosis by Targeting COL3A1 and MCL1 in Osteosarcoma. Med. Sci. Monit. 2018, 24, 8812–8821. [Google Scholar] [CrossRef]
  279. Kobayashi, E.; Hornicek, F.J.; Duan, Z. MicroRNA involvement in osteosarcoma. Sarcoma 2012, 2012, 359739. [Google Scholar] [CrossRef]
  280. Lurain, J.R.; Elfstrand, E.P. Single-agent methotrexate chemotherapy for the treatment of nonmetastatic gestational trophoblastic tumors. Am. J. Obstet. Gynecol. 1995, 172, 574–579. [Google Scholar] [CrossRef]
  281. Chapman-Davis, E.; Hoekstra, A.V.; Rademaker, A.W.; Schink, J.C.; Lurain, J.R. Treatment of nonmetastatic and metastatic low-risk gestational trophoblastic neoplasia: Factors associated with resistance to single-agent methotrexate chemotherapy. Gynecol. Oncol. 2012, 125, 572–575. [Google Scholar] [CrossRef] [PubMed]
  282. Couder, F.; Massardier, J.; You, B.; Abbas, F.; Hajri, T.; Lotz, J.-P.; Schott, A.-M.; Golfier, F. Predictive factors of relapse in low-risk gestational trophoblastic neoplasia patients successfully treated with methotrexate alone. Am. J. Obstet. Gynecol. 2016, 215, 80.e1–80.e7. [Google Scholar] [CrossRef] [PubMed]
  283. Lawrie, T.A.; Alazzam, M.; Tidy, J.; Hancock, B.W.; Osborne, R. First-line chemotherapy in low-risk gestational trophoblastic neoplasia. Cochrane Database Syst. Rev. 2016, 2016, CD007102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. May, T.; Goldstein, D.P.; Berkowitz, R.S. Current Chemotherapeutic Management of Patients with Gestational Trophoblastic Neoplasia. Chemother. Res. Pract. 2011, 2011, 12. [Google Scholar] [CrossRef] [PubMed]
  285. Aminimoghaddam, S.; Abolghasem, N.; Ashraf-Ganjooie, T. New Management of Gestational trophoblastic diseases; A Continuum of Moles to Choriocarcinoma: A Review Article. J. Obstet. Gynecol. Cancer Res. 2018, 3, 123–128. [Google Scholar]
  286. Alazzam, M.; Tidy, J.; Osborne, R.; Coleman, R.; Hancock, B.W.; Lawrie, T.A. Chemotherapy for resistant or recurrent gestational trophoblastic neoplasia. Cochrane Database Syst. Rev. 2016, 1. [Google Scholar] [CrossRef]
  287. Skubisz, M.M.; Tong, S. The Evolution of Methotrexate as a Treatment for Ectopic Pregnancy and Gestational Trophoblastic Neoplasia: A Review. Obstet. Gynecol. 2012, 2012, 8. [Google Scholar] [CrossRef]
  288. Agarwal, R.; Harding, V.; Short, D.; Fisher, R.A.; Sebire, N.J.; Harvey, R.; Patel, D.; Savage, P.M.; Lim, A.K.; Seckl, M.J. Uterine artery pulsatility index: A predictor of methotrexate resistance in gestational trophoblastic neoplasia. Br. J. Cancer 2012, 106, 1089–1094. [Google Scholar] [CrossRef] [PubMed]
  289. Seckl, M.J.; Sebire, N.J.; Fisher, R.A.; Golfier, F.; Massuger, L.; Sessa, C. Gestational trophoblastic disease: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2013, 24, vi39–vi50. [Google Scholar] [CrossRef]
  290. Hussain, A.; Aziz, S.A.; Bhat, G.M.; Lone, A.R. Management of gestational trophoblastic neoplasia. Clin. Obstet. Gynecol. Reprod. Med. 2015, 1, 5–11. [Google Scholar] [CrossRef] [Green Version]
  291. Biscaro, A.; Braga, A.; Berkowitz, R.S. Diagnosis, classification and treatment of gestational trophoblastic neoplasia. Rev. Bras. Ginecol. Obstet. 2015, 37, 42–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  292. Eysbouts, Y.K.; Ottevanger, P.B.; Massuger, L.F.A.G.; IntHout, J.; Short, D.; Harvey, R.; Kaur, B.; Sebire, N.J.; Sarwar, N.; Sweep, F.C.G.J.; et al. Can the FIGO 2000 scoring system for gestational trophoblastic neoplasia be simplified? A new retrospective analysis from a nationwide dataset. Ann. Oncol. 2017, 28, 1856–1861. [Google Scholar] [CrossRef] [PubMed]
  293. Stewart, M.; Malkovska, V.; Krishnan, J.; Lessin, L.; Barth, W.; Krishnan, J.; Lessin, L.; Barth, W. Lymphoma in a patient with rheumatoid arthritis receiving methotrexate treatment: Successful treatment with rituximab. Ann. Rheum. Dis. 2001, 60, 892–893. [Google Scholar] [PubMed]
  294. Mariette, X.; Cazals-Hatem, D.; Warszawki, J.; Liote, F.; Balandraud, N.; Sibilia, J. Lymphomas in rheumatoid arthritis patients treated with methotrexate: A 3-year prospective study in France. Blood 2002, 99, 3909–3915. [Google Scholar] [CrossRef] [Green Version]
  295. Gelfand, J.M.; Shin, D.B.; Neimann, A.L.; Wang, X.; Margolis, D.J.; Troxel, A.B. The risk of lymphoma in patients with psoriasis. J. Invest. Dermatol. 2006, 126, 2194–2201. [Google Scholar] [CrossRef] [Green Version]
  296. Jankowitz, R.C.; Ganon, J.; Blodgett, T.; Garcia, C.; Jacobs, S. A Putative Case of Methotrexate-Related Lymphoma: Clinical Course and PET/CT Findings. Case Rep Med 2009, 2009. [Google Scholar] [CrossRef]
  297. Anderson, L.A.; Gadalla, S.; Morton, L.M.; Landgren, O.; Pfeiffer, R.; Warren, J.L.; Berndt, S.I.; Ricker, W.; Parsons, R.; Engels, E.A. Population-based study of autoimmune conditions and the risk of specific lymphoid malignancies. Int. J. Cancer 2009, 125, 398–405. [Google Scholar] [CrossRef] [Green Version]
  298. Hagihara, M.; Mese, T.; Ohara, S.; Hua, J.; Ide, S.; Inoue, M. Methotrexate-associated Intravascular Large B-cell Lymphoma in a Patient with Rheumatoid Arthritis: A Very Rare Case. Intern. Med. 2018, 57, 3001–3005. [Google Scholar] [CrossRef] [Green Version]
  299. Bolonga, C.; Picot, M.C.; Jorgensen, C.; Viu, P.; Verdier, R.; Sany, J. Study of eight cases of cancer in 426 rheumatoid arthritis patients treated with methotrexate. Ann. Rheum. Dis. 1997, 56, 97–102. [Google Scholar] [CrossRef] [Green Version]
  300. Ruymann, F.B.; Mosijczuk, A.D.; Sayers, R.J. Hepatoma in a Child with Methotrexate-Induced Hepatic Fibrosis. JAMA 1977, 238, 2631–2633. [Google Scholar] [CrossRef]
  301. Al-Anazi, K.A.; Eltayeb, K.I.; Bakr, M.; Al-Mohareb, F.I. Methotrexate-induced acute leukemia: Report of three cases and review of the literature. Clin. Med. Case Rep. 2009, 2, 43–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  302. Jones, L.A.; Robertson, N.P. An update on treatments in myasthenia gravis. J. Neurol. 2017, 264, 205–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Structure of folic acid and its derivatives - aminopterin and methotrexate.
Figure 1. Structure of folic acid and its derivatives - aminopterin and methotrexate.
Ijms 21 03483 g001
Figure 2. Scheme of mechanism of methotrexate action. MTX—methotrexate; MTXGlu—polyglutamated methotrexate; FRs—folate receptors; RFC1—reduced folate carrier 1; FPGS—folylpolyglutamyl synthase; DHF—dihydrofolate; DHFR—dihydrofolate reductase; THF—tetrahydrofolate; dUMP—deoxyuridine monophosphate; TYMS—thymidylate synthase; dTMP—deoxythymidine monophosphate; DNA—deoxyribonucleic acid; RNA—ribonucleic acid; AICAR—5-aminoimidazole-4-carboxamide ribonucleotide; AICART—5-aminoimidazole-4- carboxamide ribonucleotide transformylase; FAICAR—5-formamidoimidazole-4-carboxamide ribonucleotide; IMP—inosine monophosphate; GMP—guanosine monophosphate; AMPDA—adenosine monophosphate deaminase; ADA—adenosine deaminase; AMP—adenosine monophosphate; CD73—ecto-5′-nucleotidase; A1, A2A, A2B or A3—adenosine receptors. Green arrows represent stimulation, red arrows and sticks represent inhibition, blue arrows represent biochemical conversion, yellow arrows represent migration.
Figure 2. Scheme of mechanism of methotrexate action. MTX—methotrexate; MTXGlu—polyglutamated methotrexate; FRs—folate receptors; RFC1—reduced folate carrier 1; FPGS—folylpolyglutamyl synthase; DHF—dihydrofolate; DHFR—dihydrofolate reductase; THF—tetrahydrofolate; dUMP—deoxyuridine monophosphate; TYMS—thymidylate synthase; dTMP—deoxythymidine monophosphate; DNA—deoxyribonucleic acid; RNA—ribonucleic acid; AICAR—5-aminoimidazole-4-carboxamide ribonucleotide; AICART—5-aminoimidazole-4- carboxamide ribonucleotide transformylase; FAICAR—5-formamidoimidazole-4-carboxamide ribonucleotide; IMP—inosine monophosphate; GMP—guanosine monophosphate; AMPDA—adenosine monophosphate deaminase; ADA—adenosine deaminase; AMP—adenosine monophosphate; CD73—ecto-5′-nucleotidase; A1, A2A, A2B or A3—adenosine receptors. Green arrows represent stimulation, red arrows and sticks represent inhibition, blue arrows represent biochemical conversion, yellow arrows represent migration.
Ijms 21 03483 g002
Table 1. Methotrexate application as non-oncology drug.
Table 1. Methotrexate application as non-oncology drug.
DiseaseFDA/EMA ApprovalReferences
Myasthenia gravis−/−[17,80,81,82,83,84,85,86,87,88,89,90]
Rheumatoid arthritis+/+[14,15,95,100,101,102]
Psoriasis+/+[16,68,107,114,115,116]
Crohn’s disease−/−[18,117,118]
Sarcoidosis+/+[68,114,115,116]
Alzheimer’s disease−/−[104,105,106]
Multiple sclerosis−/−[19,121,122,123]
Table 2. Methotrexate application as anti-tumour agent.
Table 2. Methotrexate application as anti-tumour agent.
Content of Combination RegimensType of CancerReference
LDMTXchildhood ALL[128]
PCNSL[129,130]
mycosis fungoides[131,132,133,134]
HNSCC[135]
LDMTX + thalidomiderefractory NHL[136,137]
LDMTX + bleomycinHNSCC[138]
LDMTX-LV + PDD + 5-FU[139]
LDMTX or LDMTX-5-FU[140]
LDMTX + celecoxib[141,142]
LDMTX + capecitabine[143]
LDMTX + interferonsmycosis fungoides[131,144,145,146,147]
LDMTX + 311 nm UV-B[148]
LDMTX + radiotherapy[149]
LDMTX + bleomycin + doxorubicin + topical nitrogen mustard[150]
LDMTX + CTX + lomustine +
etoposide or Adriamycin or vincristine
SCLC[151]
LDMTX-LV/HDMTX-LV + combination + CTX doxorubicin + vincristine or
LDMTX-LV/HDMTX-LV + etoposide + vincristine + hexamethylmelamine
[152]
LDMTX-LV/HDMTX-LV + vincristine + doxorubicin + CTX [MOAC][153]
LDMTX + PDDNSCLC[154]
LDMTX + mitomycin C + vinblastin + PDD
[M-MVP]
[155]
LDMTX-LV / HDMTX-LVbladder cancer[10,156,157,158]
LDMTX + PDD[159]
LDMTX + vinblastine + Adriamycin + cisplatin [MVAC][160,161,162,163,164,165]
HDMTX + vinblastine + Adriamycin + PDD [HDMVAC]
LDMTX + cisplatin + vinblastine [CMV]
IDMTXchildhood ALL[166]
HDMTX-LVchildhood ALL[167,168,169,170]
PCNSL, widespread NHL[171,172,173,174,175,176]
HDMTX + citrovorum factorALL (B-lineage or T-lineage)[177]
HDMTX + cytarabine NHL[178,179,180]
HDMTX + temozolomidePCNSL[181]
HDMTX + rituximab + cytarabine + dexamethasone (R-MAD)lymphoma[182]
HDMTX+ bleomycin + Adriamycin + Cytoxan + vincristine[172]
HDMTX + rituximab +CTX+ doxorubicin + vincristine + prednisolone
(R-CHOP)
[183]
HDMTX + rituximab + procarbazine + vincristine
(R-MPV)
[184]
HDMTX + rituximab[185,186,187]
HDMTX-BCG vaccineHNSCC[188]
MTX-LV + 5-FUmycosis fungoides[189]
MTX-(glutamine)2brain tumour delivery system[190]
MTX-(lysine)2[191]
MTX- magneticNPs-HSA[192,193,194,195]
MTX- magnetic(iron oxide)NPs[196]
MTX-polyacrylateNPs[197]
MTX-chitosanNPs[198]
MTX-polymersNPs[199,200]
MTX-protein microspheres[199,200,201,202,203,204,205,206]
MTX-dendrimers [G5(MTX)n][62]
MTX-dendrimers [C225-G5-MTX][206,207,208,209]
MTX-dendrimers [PEPE-MTX][210]
MTX-PEG[211]
MTX-EPR[212]
CTX + MTX + 5-FU [CMF]
5-FU + Adriamycin + CTX [FAC]
MTX-LV + tegafur and/or uracil [MUL]
breast cancer
(TNBC)
[213,214,215,216,217]
MTX + CTX[218]
[CMF] + CMFEV + epirubicin or vincristine (rotationally) [CMFEV][216]
MTX + mitomycin C [MM][219]
MTX + 5-aminoimidazole-4-carboxamide riboside (AICAR)[220]
MTX + vitamin C[221]
HDMTX-LV + mechlorethamine + procarbazineNSCLC[222]
MTX + PTX + PCNPs [MTX-PMX-PCNPs][223]
MTX + pretubulysinNSCLC (in vitro study)[224]
MTX + acetylsalicylic acid (ASA)[225]
MTX-gelatinNPspulmonary lung delivery[226]
HDMTX + 5-FU + vincristine + bleomycin + mitomycin Cblade cancer[227,228]
HDMTX-LV + doxorubicin + cisplatin + ifosfamide and/or CTXosteosarcoma[229]
HDMTX + doxorubicin + PDD [MAP]
HDMTX + doxorubicin + PDD + IFN-α-2b
[230]
HDMTX or moderate MTX + Adriamycin + vincristine [231]
HDMTX + cisplatin + isofosfamide + doxorubicin + muramyl tripeptide encapsulated in liposomes [232]
HDMTX + bleomycin + CTX + dactinomycin (BCD)[233]
HDMTX + cisplatin + doxorubicin [234,235]
HDMTX + trimetrexate;
HDMTX + PDD + doxorubicin + ifosfamide + etoposide + zoledronic acid
[236]
MTX + ACT [MACT]low risk GTN[237]
HDMTX + etoposide;
etoposide + cisplatin /+/ etoposide + MTX + dactinomycin [EP-EMA]
high risk GTN[238]

Share and Cite

MDPI and ACS Style

Koźmiński, P.; Halik, P.K.; Chesori, R.; Gniazdowska, E. Overview of Dual-Acting Drug Methotrexate in Different Neurological Diseases, Autoimmune Pathologies and Cancers. Int. J. Mol. Sci. 2020, 21, 3483. https://doi.org/10.3390/ijms21103483

AMA Style

Koźmiński P, Halik PK, Chesori R, Gniazdowska E. Overview of Dual-Acting Drug Methotrexate in Different Neurological Diseases, Autoimmune Pathologies and Cancers. International Journal of Molecular Sciences. 2020; 21(10):3483. https://doi.org/10.3390/ijms21103483

Chicago/Turabian Style

Koźmiński, Przemysław, Paweł Krzysztof Halik, Raphael Chesori, and Ewa Gniazdowska. 2020. "Overview of Dual-Acting Drug Methotrexate in Different Neurological Diseases, Autoimmune Pathologies and Cancers" International Journal of Molecular Sciences 21, no. 10: 3483. https://doi.org/10.3390/ijms21103483

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop