11,231
Views
146
CrossRef citations to date
0
Altmetric
Review

Transfersomes as versatile and flexible nano-vesicular carriers in skin cancer therapy: the state of the art

, ORCID Icon &
Article: 1325708 | Received 14 Feb 2017, Accepted 28 Apr 2017, Published online: 07 Jun 2017

ABSTRACT

Introduction: The skin acts as a barrier and prevents transcutaneous delivery of therapeutic agents. Transfersomes are novel vesicular systems that are several times more elastic than other vesicular systems. These are composed of edge activator, phospholipids, ethanol, and sodium cholate and are applied in a non-occlusive manner.

Areas covered: This article covers information such as merits/demerits of transfersomes, regulatory aspects of materials used in preparation, different methods of preparation, mechanism of action, review of clinical investigations performed, marketed preparations available, research reports, and patent reports related to transfersomes.

Expert opinion: Research over the past few years has provided a better understanding of transfersomal permeation of therapeutic agents across stratum corneum barrier. Transfersomes provides an essential feature of their application to variety of compositions in order to optimize the permeability of a range of therapeutic molecules. This is evidenced by the fact that there are several Transfersome products being processed in advanced clinical trials. It is noteworthy that a number of Transfersome products for dermal and transdermal delivery will gain a global market success in near future.

1. Introduction

​​Cancer is one of the lead factors responsible for deaths worldwide, caused by persistent tissue injury, host environment relations, etc. The frequent contact of carcinogens such as tobacco, ultraviolet light, and infections leads to various genetic (mutations), epigenetic (loss of heterozygosity), and worldwide transcriptome changes (via inflammation pathways) and is linked with increased cancer risk [Citation1]. Owing to increased rate of cancer and global prevalence during the last decade, it has posed a great challenge to health-care professionals. World Health Organization (WHO) statistics suggest a concerning 45% boost in global cancer deaths by 2030, of which 70% would be contributed from emerging countries such as India [Citation2]. With constant progression in the field of science and technology, the need to address the practical problems associated with the drug therapies increased proportionately. Cutaneous melanoma is the most violent skin cancer, accounting for 75% of all deaths [Citation3].

In 2012, the most common causes of cancer death worldwide (for both sexes) were:

  1. Lung cancer (19% of all cancer deaths; 1.6 million people).

  2. Liver cancer (9% of all cancer deaths; 745,000 people).

  3. Stomach cancer (9% of all cancer deaths; 723,000 people).

  4. Colorectal cancer (9% of all cancer deaths; 694,000 people).

  5. Breast cancer (6% of all cancer deaths; 522,000 people).

  6. Cancer of the esophagus (5% of all cancers diagnosed; 400,000 people).

  7. Pancreas cancer (4% of all cancers diagnosed; 330,000 people).

In 2012, the most common causes of cancer death worldwide (for males and females) were:

  1. Among males: lung, liver, stomach, colorectal, and prostate.

  2. Among females: breast, lung, colorectal, cervical, and stoma

During 2016, a predicted population of around 1,685,210 consisted new cases of cancer which were diagnosedin the United States out of which around 595,690 people were died from the disease.​ In 2016, the most prevailing forms of cancer were; breast cancer, lung and bronchus cancer, prostate cancer, colon and rectum cancer, bladder cancer, melanoma of the skin, Non-Hodgkin lymphoma, thyroid cancer, kidney and renal pelvis cancer, leukemia, endometrial cancer, and pancreatic cancer.[Citation4].​

1.1. Transfersomes

The newly presented novel drug carriers are highly deformable vesicles, i.e. transfersomes, which can carry large molecules across intact mammalian skin.​ A transfersome, in the widest sense of the word, is a tool that can pass instinctively through a skin and transfer drugs from the application to the target site [Citation5Citation7]. The use of lipid vesicles as a drug delivery system for skin treatment has gained increasing attention this year, but it remains controversial; mainly relevant reports cite the localization effect of liposomes, with the transport processes reported in a few cases, depending on the formulation [Citation8,Citation9]. To resolve these issues, a novel type of highly deformable lipid vesicle known as transfersome has been reported recently to go through intact skin, if applied non-occlusively, since non-occlusive conditions are essential to generate a transepidermal osmotic gradient, which acts as the driving force for elastic transport into the skin. The osmotic gradient is caused by the difference in water concentration between the skin surface and skin interiors. Transfersomes are highly deformable, and this property assists in their quick penetration through the intercellular lipid pathway of the subcutaneous tissue.​ Some of the exploratory findings reported on the existence of misdeeds inside the intercellular lipid packing of murine subcutaneous tissue, which acts as the virtual channel through which transfersomes can penetrate [Citation6,Citation10]. Transfersomes have been defined as specially designed vesicular particles consisting of at least one inner aqueous compartment enclosed by lipid vesicles; liposomes in morphology, but, functionally, transfersomes are suitably deformable to go through pores much smaller than their own size. A schematic diagram of the structure of transfersomes is presented in .

Figure 1. Structure of transfersomes.

Figure 1. Structure of transfersomes.

Introduced in 1992, Cevc and Blume second-generation vesicular carriers, named Ultradeformable liposomes or Transfersomes®, possess slighter vesicular size (typically <300 nm) and higher elasticity (typically five–eight times higher compared with conventional liposomes) [Citation10Citation12].

1.2. Historical background

The transfersome term was first introduced by Cevc [Citation10] and has been the subject of several patents and literature information since the 1990s (Transfersomes, a trademark of IDEA AG, Munich, Germany), and it represents the first generation of ultradeformable vesicles. The skin permeation and penetration of these elastic vesicles result from a synergic mechanism among the carrier properties and the access enrichment ability.​ Transfersomes are ultradeformable lipid bundles of aggregates in supramolecular form constructed with a minimum of one interior aqueous segment encircled by a lipid bilayer exhibiting adapted properties, which are appropriate under the presence of surfactants in the vesicular membrane (edge activator (EA)) [Citation13Citation18].​ Even if it is generally accepted that the permeation of, usually, liposomes is limited to the outer layer of the stratum corneum, thus providing a drug or cosmetic localizing effect within the skin, transfersomes are claimed to infuse as intact vesicles through the skin layers to the complete circulation. Non-steroidal anti-inflammatory drug (NSAID) ketoprofen was successful on a validation basis and was immensely popular in the market. Ketoprofen was authorized by a Swiss regulatory agency (Swiss Medic) in 2007; the trade name was ‘Ketoprofen transdermal’, manufactured by ‘IDEA AG’ pharmaceuticals Pvt. Ltd.

2. Merits/demerits

Transfersomes defeat the skin obstacle by opening extracellular pathways among the cells in the organ and then deforming to fit into such passages. In the process, transfersomes go through a series of stress-dependent adjustments of the local carrier symphony to minimize the struggle of motion through the otherwise confining channels.​ This process allows transfersomes to convey the drugs associated into and diagonally across the skin easily and very reproducibly.​ This happens at a rate largely higher than that achieved by more predictable formulations and offers an excellent means for controlling drug distribution in the skin [Citation19,Citation20]. Transfersomes have been used as carriers for different therapeutic agents, including proteins, insulin [Citation21,Citation22], DNA [Citation23], gap junction protein [Citation24], peptides [Citation25], albumin [Citation15], nutraceuticals [Citation26], corticosteroids [Citation27], antigens [Citation28], analgesics [Citation29], sex hormones [Citation30], and anesthetics [Citation31], and have been proven to augment significantly the amount of drug permeated through the skin [Citation10]. The topical application of transfersome-entrapped anticancer drug is described by a few research groups. Skin delivery of 5-fluorouracil (5-FU) [Citation32,Citation33,Citation34], methotrexate, and bleomycin has been evaluated by many researchers. Edge activators are often single chain surfactant that destabilizes lipid bilayers of the vesicles and provides a flexible membrane, eventually making transfersomes highly flexible. The role of sodium deoxycholate, Spans, and Tween on the skin penetration and deposition is discussed in [Citation35Citation37]. The application of transfersomes does not engage any intricate procedure and they can be applied by a non-occluded process, whereby they pass throughout the multilayered lipid matrix of the stratum corneum as an outcome of the hydration or osmotic force within the skin [Citation38]. One main drawback of these vesicles corresponds to the difficulty of loading hydrophobic drugs into the vesicles without compromising their deformability and elastic properties [Citation39]. Transfersomes, by virtue of their enhanced elasticity in contrast to standard liposomes, are more amenable to the transport of therapeutic agents across the human skin. [Citation40].​

2.1. Mechanism of action

The present investigation indicates that the transfersomes are drug mover systems that can penetrate across intact skin. It is believed that the unimpeded passage of such carriers is based on two key factors: the high elasticity (deformability) of the vesicle bilayers and the reality of an osmotic gradient across the skin. Because of their high deformability, transfersomes with the help of EAs generate a transepidermal osmotic gradient; and further squeeze among the stratum corneum cells and carry drug across the whole skin [Citation10]. The transpore hydrostatic force difference is liable for the penetration or passage of transfersomes intact throughout the stratum corneum, i.e. the penetration of transfersome is an outcome of hydrotaxis and the permeation is governed by principles of elastomechanics [Citation41]. When a transfersome reaches a pore, it is capable of changing its membrane work reversibly as an effect of its self-optimizing deformability. To go throughout the pore, the mechanism of the transfersome liable for its deformability starts accumulating at the site of tension, whereas the less elastic mechanism experiences dilution, which significantly reduces the active rate of membrane deformation and allows the highly elastic particles to go throughout the pores.​ The passage of transfersomes through the skin and the epithelial obstacle is greatly prejudiced by the flexibility of their membrane, which can be achieved via a suitable ratio of surfactants. The flexibility of the transfersomal membrane decreases the risk of complete vesicle rupture in the skin and permits the ultradeformable transfersomes to change their membrane composition locally and reversibly when they are pressed against or attracted into a narrow pore. This dramatically lowers the energetic cost of membrane deformation and permits the resulting highly flexible particles first to enter and then to pass through the pores rapidly and efficiently [Citation42]. A schematic diagram of the mechanism of action of transfersomes is presented in .

Figure 2. Schematic presentation of mechanism of action of transfersome in transdermal delivery of anticancer agent.

Figure 2. Schematic presentation of mechanism of action of transfersome in transdermal delivery of anticancer agent.

The first mechanism proposes that vesicles can act as drug carrier systems, whereby intact vesicles enter the stratum corneum carrying vesicle-bound drug molecules into the skin under the influence of the naturally occurring in vivo transcutaneous hydration gradient [Citation13].

3. Regulatory aspects

Recently, advances in pharmaceutical science and skill have made available a range of new excipients, such as lipids, surfactants, and solvents; though, of late, there have been reservations within the scientific community regarding the dullness of excipients and that they in some capacity have unfavorable effects. Selection of an excipient throughout the research of a transfersome-based formulation is limited by safety and toxicity concerns associated with these excipients. Hence, a small range of excipients are obtainable for planning any highly porous drug deliverance system. Thus, inert excipients are usually measured when developing a transfersome-based formulation and these are used as vesicle-forming agents, surfactants, EAs, and solvents. Mitigating the safety concerns, a narrow range of excipients are obtainable for crafting any highly porous drug delivery system, such as a transfersome [Citation43]. Different national regulatory agencies (WHO, International Pharmaceutical Excipients Council, US Food and Drug Administration (FDA), Japanese Ministry of Health and Welfare, and International Conference on Harmonisation of Technical necessities for muster of Pharmaceuticals for Human Use) have maintained a confidential list of excipients as ‘Generally Regarded as Safe’ (GRAS), which have been clinically categorized not to be toxic. The FDA keeps a record entitled ‘Inactive Ingredient Guide’, which includes a catalogue of permitted excipients. This documentation provides information about the excipients with a value of their utmost dosage stage by a fastidious route of direction or dosage form [Citation44]. Phospholipid is a crucial element for the formation of a transfersome-based drug delivery system. It is also roughly always true that the fluid-chain vesicles with a rather elastic bilayer promote drug transport across skin obstruction better than the more rigid liposomes [Citation45]. Therefore, nearly all the common phosphatidylcholine (PC) used to organize stretchy liposomes is unsaturated PC (i.e. soybean phosphatidylcholine (SPC) or egg phosphatidylcholine (EPC)). SPC is a GRAS-listed phospholipid and also complies with specifications of the Food Chemicals Codex (http://www.NutriScienceUSA.com). Edge activator is generally a kind of surfactant which destabilizes the lipid bilayer of the elastic liposomes and increases elasticity of the bilayer concurrently. Amid EAs, sodium cholate, sodium deoxycholate, Span-80, Tween-80 and Tween-20 were normally used. ​ Biju et al. recommended that some chemical penetration enhancers such as oleic acid can be used as well as EA to replace the normally used surfactant [Citation14]. The survival of mixed micelles also leads to lower drug trap due to their higher inflexibility and smaller size [Citation46,Citation47].

Edge activator plays an important role in determining the skin permeation behavior of elastic liposomes. An overview of the differences among EAs is helpful for the selection of an ideal EA for optimal formulation. Sodium deoxycholate is a water-soluble ionic surfactant. Valsartan-loaded elastic liposomes containing sodium deoxycholate as the EA were then investigated [Citation48]. Similarly, sodium cholate, which is used as an EA, is reported to be non-toxic but has been kept in the hazardous category as it causes skin and eye irritations as well as respiratory sensitization. Surfactants can cause severe gastrointestinal discomfort when used above certain concentrations; the maximum safe limit of surfactant concentration is 10–25%. Ethanol is known to act as an efficient skin-penetration enhancer. It can interact with the polar head group region of the lipid molecules, resulting in a reduction of the melting point of the stratum corneum lipids, thereby increasing their fluidity and cell membrane permeability.

4. Method of preparation

4.1. Rotary film evaporation method

This method is also known as the hand-shaking process, which was initially invented by Bangham [Citation49]. In this process, the quantity needed of phospholipids and surfactants (as EAs) is essential to organize a thin film [Citation50,Citation51].​ It is largely worn for the research of multilamellar vesicles. A solution of phospholipids and EAs is organized in a crude solvent such as a combination of chloroform and methanol. The prepared solution is transferred to a round-bottomed flask which is rotated at constant temperature (above the glass transition temperature of lipids) and reduced pressure. A film of lipids and EA is formed on the walls of the flask. The twisted film is then hydrated using aqueous media containing drug. This causes lipids to swell and form bilayer vesicles. Vesicles of desired size can be obtained by extrusion or by sonication of the superior vesicles [Citation52].

4.2. Reverse-phase evaporation method

At this point, the scheme will alter to a viscous gel followed by the arrangement of vesicles. The non-encapsulated material and residual solvents can be indifferentiable using dialysis or centrifugation [Citation52].​ In this method, lipids dissolved in organic solvents are collected in a round-bottomed flask. Aqueous media containing EAs is added under nitrogen purging. The drug can be added to the lipid or aqueous medium based on its solubility character. The system formed is then sonicated, awaiting its conversion into a standardized dispersion, and should not separate for at least 30 min after sonication. The organic solvent is then removed under low pressure.​

4.3. Vortexing sonication method

In the vortexing sonication method, mixed lipids (i.e. phosphatidylcholine, EA and the therapeutic agent) are blended in a phosphate buffer and vortexed to attain a milky suspension. The suspension is sonicated, followed by extrusion through polycarbonate membranes [Citation53]. Cationic transfersomes have also been set by this method, which involves mixing cationic lipids, such as DOTMA, with PBS to attain a concentration of 10 mg/ml followed by a count of sodium deoxycholate (SDC). The blend is vortexed and sonicated, followed by extrusion through a polycarbonate (100 nm) filter.

4.4. Ethanol injection method

In this process, the aqueous solution containing drug is heated with unremitting stirring at constant temperature. Ethanolic solution of phospholipids and EAs is injected into aqueous solution dropwise. As the solution comes into contact with aqueous media the lipid molecules are precipitated and form bilayered structures. This process offers assorted advantages over other methods, which include simplicity, reproducibility, and scale-up [Citation54,Citation55].

4.5. Freeze–thaw method

This method includes the exposure of multilamellar vesicles to alternate cycles of very low temperature for freezing followed by exposure to very high temperature.​ The geared-up suspension is transferred to a tube and dipped in a nitrogen bath (−30°C) for 30 s. After freezing, it is exposed to a high temperature in a water bath. This course is repeated eight–nine times [Citation56]. An application of transfersomes in the delivery of various therapeutic agents is summarized in .

Table 1. Examples of research reports on using transfersomes as carriers for the delivery of therapeutic agents.

5. Applications

5.1. Actinic keratosis

Actinic keratosis (AK) is an ordinary skin ailment caused by long-standing sun exposure, and classically forms on the face, neck, balding scalp, chest, shoulders, and the back of arms and hands of adults, 75% of all reported lesions existing on the head, neck, and forearms. Actinic keratosis is characterized by the shape of keratotic macules, papules, or plaques with superficial scales on a red base. Lesions are frequently asymptomatic, but they can be painful or itchy. Owing to the swelling nature of the condition, the occurrence of AK increases with age and is an ordinary condition in the adult populace aged over 50 years [Citation84]. Treatment of AK depends on the medical appearance of the lesions: it may be fought at exact lesions (lesion directed) or at numerous lesions over a large area (field directed), and occasionally both treatment approaches are used. Therapy options include cryosurgery, curettage, excision surgery, photodynamic therapy (PDT), and topical treatments (5-FU cream, diclofenac gel, imiquimod cream, and ingenol mebutate gel).

A study in England reported a prevalence rate of 15.4 and 5.9% in men and women, respectively. This rate amplified to 34.1 and 18.2% in men and women aged above 70 years [Citation85]. The study set up to investigate the occurrence of AK was better in individuals with red hair and freckles, which indicates Fitzpatrick skin type I.​ There was a similar, but extra-marked, boost reported in Australia; prevalence rates of AK were 22 and 8% for men and women aged 30–39 years, which increased to 83 and 64%, respectively, in adults aged 60–69 years [Citation86]. One reason for the greater incidence rate in males might be that it is more possible for them to labor outdoors and have more sun contact.

5.1.1. Topical delivery of 5-FU for the treatment of AK and non-melanoma skin cancer

​ Unfortunately, 5-FU showed poor percutaneous permeation, thus reducing its anticancer effectiveness after topical administration. The in vivo results concluded that vesiculization of 5-FU not only improves the topical delivery, but also enhances the cytotoxic effect of 5-FU [Citation87]. An instance of transfersomal gel containing 5-FU provided efficient results against the treatment for AK and non-melanoma skin carcinoma, which showed up to a twofold increase of transdermal release in contrast to other marketed formulations [Citation88].

5.2. Basal cell carcinoma

The frequency of basal cell carcinoma shows clear environmental variation. The age-standardized occurrence of basal cell carcinoma in South Wales was predictable at 114 per 100,000 population in 1998 [Citation89]. The common age and sex-standardized annual occurrence in Minnesota, USA, was reported at 146 per 100,000. In Australia, the rate is ahead at 726 per 100,000 [Citation90]. These statistics are likely to be underestimates, as basal cell carcinoma tends to be under-reported to the cancer registries. In white populations in North America, the frequency has improved more than 10% a year, foremost to a lifetime risk of 30% of growing a basal cell carcinoma [Citation91]. With an ever increasing elderly population, the disease is likely to become more of a problem in the future. Certainly, the prevalence of this cancer will probably be greater than that of all other cancers mutually. The age-standardized occurrence of basal cell carcinoma in white populations is generally between 18 and 40% higher in men (British and Australian data) [Citation92]. Basal cell carcinoma is extremely uncommon in dark-skinned races. Skin type I (always burns, never tans), red or pale hair, and blue or green eyes have been shown to be risk factors for the development of basal cell carcinoma, with an estimated odds ratio of 1.6 [Citation93]. Advance of basal cell carcinoma is reported to be more frequent after freckling in childhood and also after frequent or severe sunburn in childhood [Citation94,Citation95]. This is in contrast to a history of sunburn as an adult, which does not seem to be associated with the development of basal cell carcinoma [Citation95].

Other non-ultraviolet ecological exposures that have been connected with amplified risk of basal cell carcinoma include ionizing radiation, high dietary energy (especially fat), low intake of vitamins, and various chemicals and dust. Introduction to arsenic predisposes one to multiple basal cell carcinomas [Citation96,Citation97].

Patients on an immunosuppressive cure also have an increased risk of basal cell carcinoma. A study in the Netherlands showed that the occurrence of basal cell carcinoma in transplant recipients was 10 times higher than in the general population [Citation97].​

Fadel et al. [Citation98] reported that indocyanine green was encapsulated in a vesicular colloidal nanocarrier (transfersomes) for potential application as a photosensitizer in topical PDT of basal cell carcinoma.

5.3. Kaposi’s sarcoma

Kaposi’s sarcoma (KS) was first described in 1872 by the Hungarian dermatologist Moritz Kaposi. At that time, before human immunodeficiency virus (HIV) and acquired immune deficiency syndrome (AIDS), KS remained a rare tumor.​ While the majority of the cases seen in Europe and North America have occurred in aged men of Italian or Eastern European Jewish ancestry, the neoplasm also occurs in some other distinct populations: young black African adult males, prepubescent children, renal allograft recipients, and other patients receiving immunosuppressive therapy. The disseminated, fulminate form of KS associated with HIV disease is referred to as epidemic KS to distinguish it from the classic, African, and transplant-related varieties of the neoplasm. In addition, KS has been acknowledged in homosexual men apart from the HIV disease epidemic [Citation99]. In the 1950s, KS was known as a comparatively ordinary neoplasm endemic in inhabitant populations in equatorial Africa and comprised approximately 9% of all cancers seen in Ugandan males. During 1969, the first case of KS in connection with immunosuppression in a renal transplant patient was described. At that time, a number of renal and additional organ allograft recipients who received conventional prednisone and azathioprine developed KS shortly after the onset of immunosuppressive therapy [Citation100]. Pathak et al. [Citation101]​ developed deformable nanovesicles of paclitaxel capable of being used in dermal chemotherapy, especially deep into the dermal areas of AIDS-related KS. An in vitro cytotoxicity study on KSY-1 cell lines revealed higher IC50 (≤17) for transfersome against IC50 ≤ 19 for transfersome gel. Confocal laser scanning microscopy confirmed the penetrating potential of transfersomes via transfersome gel to the dermal layers of skin, the proposed target site [Citation101].​

5.4. Melanoma

A melanoma is an atrocious tumor that arises from melanocytes, dendritic cells that create melanin, a tincture that protects the body from destructive ultraviolet (UV) radiation. Melanocytes use tyrosine to synthesize melanin. A cluster of melanocytes form nevi (pigmented lesions or moles), and melanoma results when these melanocytes undergo a malignant transformation. Melanocytes may be created in various areas of the body; nevertheless, they are mainly situated in the epidermis, and more than 90% of all melanomas have cutaneous incidence rates and epidemiology.​ The frequency of melanoma varies, with the peak rates in Northern Europe, New Zealand, Australia, and North America [Citation102,Citation103]. Melanoma has one of the fastest-growing rates in the US. The US occurrence rate increased from 7.9 to 17.7 per 100,000 persons between 1975 and 2000. The American Cancer Society estimated that, in 2012, about 76,250 new melanoma cases (44,250 men, 32,000 women) would be diagnosed and about 9,180 individuals (6,060 men, 3,120 women) would die from the disease [Citation104].​ Lin et al. [Citation105] formulated 5-aminolevulinic acid (5-ALA)-loaded DPPC liposomes in melanoma treatment. The results found that 5-ALA/DPPC formulation reduced cell viability and mitochondria membrane potential, and enhanced intracellular ROS accumulation as compared with 5-ALA alone in melanoma cells. Furthermore, the 5-ALA/DPPC formulation also had better skin penetration ability as compared with the 5-ALA in our ex vivo data by assaying 5-ALA converted into protoporphyrin IX (PpIX) in the skin of the mice that were experimented on. In melanoma xenograft models, 5-ALA/DPPC enhanced PpIX accumulation only in tumor tissue, not normal skin [Citation105]. siRNAs have potential therapeutic applications in various dermatological diseases such as psoriasis, atopic dermatitis, and cancer. Dorrani et al. [Citation106] prepared a series of liposome compositions that contained various concentrations of EA in their structures and then complexed them with siRNA at different ratios to generate a small library of liposome–siRNA complexes (lipoplexes) with different physicochemical properties. Quantitative imaging analysis showed effective permeation of lipoplexes through the skin layers and deposition at the upper dermis. The ability of the formulated lipoplexes to internalize into melanoma cells, knockdown the expression of the B-raf murine sarcoma viral oncogene homolog B1 (BRAF) protein, and induce cell death in melanoma cells was studied by fluorescent microscopy, in-cell immunofluorescence assay, and WST-1 cell proliferation assay [Citation106].

5.5. Squamous cell carcinoma

Squamous cell carcinoma (SCC) is an epithelial malignancy that occurs in organs that are usually enclosed with squamous epithelium which includes numerous diverse anatomic sites, counting the skin, lips, mouth, esophagus, urinary tract, prostate, lungs, vagina, and cervix. SCC represents the most common cancer capable of metastatic spread in the US and worldwide [Citation107]. Tobacco smoking and human papilloma virus (HPV) are carcinogenic causes for all four sub-types. In total, quite a lot of risk factors are public amid the chief SCC types. Fair-skinned persons who burn and never tan are at a much higher risk for on-the-rise skin SCC than those with darker skin, and it has been confirmed that both past sun exposure and strong sun exposure appear to heavily predispose the population to skin cancer [Citation108,Citation109].

Furthermore, HPV may be involved in the multistep procedure of skin carcinogenesis as a co-factor with UV radiation, especially in patients with poor resistance, such as limb transplant recipients and smoking tobacco, which may double the risk of skin cancer [Citation110,Citation111].

Gupta et al. [Citation112] designed protransfersome for local delivery of cisplatin in cutaneous epithelial malignancies. The presence of a fluorescence marker in the skin showed better skin penetration ability of the protransfersome. The results of in vivo performance of the system showed an increase in the therapeutic efficacy of the drug with less systemic toxicity [Citation112]. Applications of transfersomes in the delivery of various anticancer agents are presented in . In addition to all this information, studies, and roles, a number of patents have been filed and granted, brief details of which, representing various formulation aspects and potential of transfersomes, are summarized in .

Table 2. Examples of research reports on using transfersomes as carriers for the delivery of anticancer agents.

Table 3. Examples of patent reports on transfersome drug delivery systems.

6. Concluding remarks

Transfersomes are personally designed vesicles capable of responding to external stress by squeezing themselves through skin pores that are many times narrower than they are, leading to increased transdermal flux of the therapeutic agents. It is clear that transfersomes can deliver enhanced amounts of both small and large therapeutic agents into and through the skin. The exact mechanism by which transport occurs remains to be elucidated, and evidence for transport of intact vesicles beyond the stratum corneum is lacking. There are increasing applications of enhanced delivery by transfersome formulations. However, there are only two transfersome-based formulations currently available in the market and the reported clinical studies mainly involve ketoprofen and insulin. The application of transfersomes in cancer presents treatment of AKs, basal cell carcinoma, SCC, melanoma, and KS. The transfersomes can be a good carrier option for delivering the drug into the skin layers and hence useful for the treatment of skin cancers. It is likely that a number of transfersome products for dermal and transdermal applications will be developed in the future.

Disclosure statement

No potential conflict of interest was reported by the authors.​

Additional information

Notes on contributors

Shubhra Rai

Ms. Shubhra Rai is a dynamic researcher and a professional academician in the field of pharmacy. She has acquired the degree of master of pharmacy in pharmaceutics. She has recently completed two national level research projects; first one as junior research fellow in “Drug to inflamed colon by polysaccharides Nanoparticals”, granted by Department of Science & Technology (DST) and second one as senior research fellow on “Guar gum montmorillonite nanocomposites for the target delivery of 5 aminosalicylic acid for management of Ulcerative colitis.” granted again by Department of Science & Technology under women empowerment scheme. Ms. Rai has also successfully completed various training sessions on “Sophisticated Analytical Instruments”. Her current area of thrust is exploring the potential of transfersomes in the delivery of therapeutic bioactives.

Vikas Pandey

Mr. Vikas Pandey is the Associate Professor of pharmaceutics and In-charge at “Drug Delivery & Nanotechnology laboratory, Department of Pharmaceutics, GRKIST college of Pharmacy. He is registered pharmacist at Madhya Pradesh State Pharmacy Council, completed his Master of Pharmacy degree from department of pharmaceutics Annamalai University, South India. He is also life member of Association of pharmaceutical teachers of India (APTI) and Indian Pharmaceutical Association (IPA). Various prevailing skin diseases in today's era gained his interest to pursue research and explore the hidden potentials of transdermal drug delivery systems. As a result he then started his PhD research work and his current area of interest is utility of transfersomes in transdermal drug delivery. He is serving pharmacy academics from last one decade and has published more than 20 international/national peer reviewed articles along with two books.

Gopal Rai

Dr. Rai is the Professor of pharmaceutics and Head at college of Pharmacy, GRKIST situated at central India. He is fellow of Association of Pharmaceutical teachers of India (APTI) and life member of Indian Pharmaceutical Association (IPA). His current project has led to the successful development of “Polysaccharide based Novel Drug Delivery System for Colon Targeting of Anticancer Drugs“ and “Polysaccharides Nanoparticles in the delivery of therapeutic bioactives to Inflamed Colon” the projects were funded by Madhya Pradesh Council of Science & Technology (MPCST), Department of Science & Technology (DST), India. Dr. Rai is serving pharmacy academics over last more than 15 years and has published more than 40 national and international peer reviewed articles. Dr. Rai has awarded with best oral presentation awards in various national and international conferences. His current area of interest is enhanced transdermal delivery of anti-cancer agents, colon drug delivery system and novel drug delivery system.

References

  • Lieu CH, William WN, Lippman SM. Cancer chemoprevention. In: Hidalgo M, Eckhardt SG, Garrett-Mayer E, et al., editors. Principles of anticancer drug development. New York: Springer Science; 2010. p. 1–18. ​
  • WHO. Cancer-fact sheets. WHO; [ cited 2017 Jan 2]. Available from: http://www.who.int/mediacentre/factsheets/fs297/en/index.html
  • Sabitha M, Sanoj Rejinold NS, Nair A, et al. Development and evaluation of 5-fluorouracil loaded chitin nanogels for treatment of skin cancer. Carbohydr Polym. 2013;91:48–57.
  • Smith RA, Andrews K, Brooks D, et al. Cancer screening in the United States, 2016: a review of current American Cancer Society guidelines and current issues in cancer screening. CA Cancer J Clin. 2016;62:95–102.
  • Cevc G. Material transport across permeability barriers by means of lipid vesicles. In: Lipowsky R, editor. Handbook of physics of biological systems. Vol. I. Vol. 9. Amsterdam: Elsevier Science; 1994. p. 441–466.
  • Cevc G, Blume G, Schatzlein A, et al. The skin: a pathway for the systemic treatment with patches and lipid-based agent carriers. Adv Drug Deliv Rev. 1996;18:349–378.
  • Cevc G, Schatzleinand A, Blume G. Transdermal drug carriers: basic properties, optimization and transfer-efficiency in the case of epicutaneously applied peptides. J Control Release. 1995;36:3–16.
  • Mezei M, Gulasekharn V. Liposomes a selective drug delivery system for topical route of administration, 1. Lotion dosages form. Life Sci. 1980;26:1473–1477.
  • Fresta M, Puglisi G. Application of liposomes as potential cutaneous drug delivery system: in vitro in vivo investigation with radioactivity labeled vesicles. J Drug Target. 1996;4:95–101.
  • Cevc G, Blume G. Lipid vesicles penetrate into skin owing to the transdermal osmotic gradients and hydration force. Biochim Biophys Acta. 1992;1104:226–232.
  • Song CK, Balakrishnan P, Shim CK, et al. A novel vesicular carrier, transethosome, for enhanced skin delivery of voriconazole: characterization and in vitro/in vivo evaluation. Colloids Surf B Biointerfaces. 2012;92:299–304.
  • Shashank J, Niketkumar P, Mansi KS, et al. Recent advances in lipid-based vesicles and particulate carriers for topical and transdermal application. J Pharm Sci. 2016;116:1–23.
  • Cevc G, Blume G. New highly efficient formulation of diclofenac for the topical, transdermal administration in ultradeformable drug carriers, transfersomes. Biochimica Et Biophysica Acta. 2001;1514(2):191–205.
  • Biju SS, Talegaonkar S, Mishra PR, et al. Vesicular system an overview. ‎Indian J Pharm Sci. 2006;68:141–153.
  • Dayan N, Touitou E. Carriers for skin delivery of Trihexiphenidyl HCl ethosomes vs liposomes. Biomaterials. 2000;21:1879–1885.
  • Hofer C, Hartung R, Gobel R, et al. New ultradeformable drug carriers for potential transdermal application of interleukin-2 and interferon-alpha theoretic and practical aspects. World J Surg. 2000;24:1187–1189.
  • Manosroi A, Jantrawuta P, Manosroi J. Anti-inflammatory activity of gel containing novel elastic niosomes entrapped with diclofenac diethylammonium. Int J Pharm. 2008;360:156–163.
  • Zheng W, Fang Xia Wang L, Zhang Y. Preparation and quality assessment of itraconazole transfersomes. Int J Pharm. 2012;436:291–298.
  • Cevc G, Blume G. Biological activity and characteristics of triamcinolone-acetonide formulated with the self-regulating drug carriers, transfersome. Biochim Biophys Acta. 2003;1614:156–164.
  • Ahad A, Aqil M, Kohli K, et al. Formulation and optimization of nanotransfersomes using experimental design technique for accentuated transdermal delivery of valsartan. Nanomed Nanotechnol Biol Med. 2012;8:237–249.
  • Jain S, Mishra D, Kuksal A, et al. Vesicular approach for drug delivery into or across the skin: current status and future prospects. Pharm Online. 2006;1:1–32.
  • Morrow DIJ, McCarron PA, Woolfson AD, et al. Innovative strategies for enhancing topical and transdermal drug delivery. Open Drug Deliv J. 2007;1:36–59.
  • Zaafarany GME, Awad GAS, Holayel SM, et al. Role of edge activators and surface charge in developing ultradeformable vesicles with enhanced skin delivery. Int J Pharm. 2010;397:164–172.
  • Jain S, Jain P. Transferosomes: a novel vesicular carrier for enhanced transdermal delivery: development, characterization and performance evaluation. Drug Dev Ind Pharm. 2003;29:1013–1026.
  • Bhardwaj V, Shukla V, Singh A, et al. Transferosomes ultra flexible vesicles for transdermal delivery. Int J Pharm Sci Res. 2010;2:12–20.
  • Schatzlein A, Cevc G. Non-uniform cellular packing of the stratum corneum and permeability barrier function of intact skin: a high resolution confocal laser scanning microscopy study using highly deformable vesicles (transfersomes). Brit J Dermatol. 1998;138:583–592.
  • Planas ME, Gonzalez P, Rodriguez L, et al. Noninvasive percutaneous induction of topical analgesia by a new type of drug carrier, and prolongation of local pain insensitivity by anesthetic liposomes. Anesth Analg. 1992;75:615–621.
  • Mahor S, Rawat A, Dubey PK, et al. Cationic transfersomes based topical genetic vaccine against hepatitis B. Int J Pharm. 2007;340:13–19.
  • Choi MJ, Maibach HI. Elastic vesicles as topical/transdermal drug delivery system. Int J Cosm Sci. 2005;27:211–221.
  • Guo J, Ping Q, Sun G, et al. Lecithin vesicular carriers for transdermal delivery of cyclosporin A. Int J Pharm. 2000;194:201–207.
  • Paul A, Cevc G, Bachhawat BK. Transdermal immunisation with an integral membrane component, gap junction protein, by means of ultradeformable drug carriers, transfersomes. Vaccine. 1998;16:188–195.
  • Gmm EM, Williams AC, Barry BW. Skin delivery of 5-fluorouracil from ultradeformable and standard liposomes in-vitro. J Pharma Pharmacol. 2001;53:1069–1077.
  • Costa CAM, Moraes AM. Encapsulation of 5-fluorouracil in liposomes for topical administration. Acta Sci Tech. 2003;25:53–61.
  • Alvi IA, Madan J, Kaushik D, et al. Comparative study of transfersomes, liposomes, and niosomes for topical delivery of 5-fluorouracil to skin cancer cells: preparation, characterization, in-vitro release, and cytotoxicity analysis. Anticancer Drugs. 2011;8:774–782.
  • Ita KB, Du Preez J, Lane ME, et al. Dermal delivery of selected hydrophilic drugs from elastic liposomes: effect of phospholipid formulation and surfactants. J Pharma Pharmacol. 2007;59:1215–1222.
  • El Zaafarany GM, Awad GAS, Holayel SM, et al. Role of edge activators and surface charge in developing ultradeformable vesicles with enhanced skin delivery. Int J Pharm. 2010;397:164–172.
  • Badran M, Shalaby K, Al-Omrani A. Influence of the flexible liposomes on the skin deposition of a hydrophilic model drug, carboxyfluorescein: dependency on their composition. Sci World J. 2012;2012:1–9.
  • Cevc G, Blume G, Schatzlein A. Transferosomes-mediated transepidermal delivery improves the regiospecificity and biological activity of corticosteroids in vivo. J Control Release. 1997;45:211–226.
  • Chen J, Lu WL, Gu W, et al. Skin permeation behavior of elastic liposomes: role of formulation ingredients. Expert Opin Drug Deliv. 2013;10:845–856.
  • Dragicevic-Curic N, Gräfe S, Gitter B, et al. Surface charged temoporfin-loaded flexible vesicles: in vitro skin penetration studies and stability. Int J Pharm. 2010;384:100–108.
  • Dubey V, Mishra D, Asthana A, et al. Transdermal delivery of a pineal hormone: melatonin via elastic liposomes. Biomaterials. 2006;27:3491–3496.
  • Myschik J, Rades T, Hook S. Advances in lipid based subunit vaccine formulations. Curr Immunol Rev. 2009;5:42–48.
  • Kumar R, Philip A. Modified transdermal technologies: breaking the barriers of drug permeation via the skin. Tropic J Pharm Res. 2007;6:633–644.
  • Owens DR. New horizons – alternative routes for insulin therapy. Nat Rev. 2002;1:529–540.
  • Cevc G, Vierl U. Nanotechnology and the transdermal route: a state of the art review and critical appraisal. J Control Release. 2010;141:277–299.
  • Cevc G. Transdermal drug delivery of insulin with ultradeformable carriers. Clin Pharmacokinet. 2003;42:461–474.
  • Kumar R, Philip A. Modified transdermal technologies: breaking the barriers of drug permeation via the skin. Tropic J Pharm Res. 2007;6:633–644.
  • Maurya SD. Enhanced transdermal delivery of Indinavir sulphate via transfersomes. Int J Comp Pharm. 2010;1:1–7.
  • Bangham AD, Standish MM, Watkins JC. Diffusion of univalent ions across the lamellae of swollen phospholipids. J Mol Biol. 1965;13:238–252.
  • Singh H, Utreja P, Tiwary A, et al. Elastic liposomal formulation for sustained delivery of colchicine: in vitro characterization and in vivo evaluation of anti-gout activity. Aaps J. 2009;11:54–64.
  • Abdallah M. Transferosomes as a transdermal drug delivery system for enhancement the antifungal activity of nyastatin. Int J Pharm Pharm Sci. 2013;5:560–567.
  • Sazoka F, Papahadjopoulos D. Procedure for preparation of liposomes with large internal aqueous space and high capture by reverse-phase evaporation. Proc Natl Acad Sci. 1978;75:4194–4198.
  • Rai K. Transfersomes: self-optimizing carriers for bioactives. PDA J Pharm Sci Technol. 2008;62:362–379.
  • Kumar A, Adde S, Kamble R, et al. Development and characterization of liposomal drug delivery system for nimesulide. Int J Pharm Pharm Sci. 2010;2:87–89.
  • Charcosset C, Juban A, Valour J, et al. Preperation of liposomes at large scale using ethanol injection method: effect of scale up and injection devices. Chem Eng Res Des. 2015;94:508–515.
  • Maestrelli F, Rodriguez M, Rabasco A, et al. Effect of preparation techniques on the properties of liposomes encapsulating ketoprofen- cyclodextrine complexes aimed for transdermal delivery. Int J Pharm. 2006;312:53–60.
  • Zhang Y, Ng W, Feng X, et al. Lipid vesicular nanocarrier: quick encapsulation efficiency determination and transcutaneous application. Int J Pharm. 2017 10;516:225–230.
  • Schlich M, Lai F, Murgia S, et al. Needle-free jet injection of intact phospholipid vesicles across the skin: a feasibility study. Biomed Microdevices. 2016;18:67.
  • Meng S, Zhang C, Shi W, et al. Preparation of osthole-loaded nano-vesicles for skin delivery: characterization, in vitro skin permeation and preliminary in vivo pharmacokinetic studies. Eur J Pharm Sci. 2016;92:49–54.
  • Hassanpour AM, Ghanbarzadeh S, Javadzadeh Y, et al. Aggregated nanotransfersomal dry powder inhalation of itraconazole for pulmonary drug delivery. Adv Pharm Bull. 2016;6:57–64.
  • González-Rodríguez ML, Arroyo CM, Cózar-Bernal MJ, et al. Deformability properties of timolol-loaded transfersomes based on the extrusion mechanism, statistical optimization of the process. Drug Dev Ind Pharm. 2016;42:1683–1694.
  • Garg V, Singh H, Bhatia A, et al. Systematic development of transethosomal gel system of piroxicam: formulation optimization, in vitro evaluation, and ex vivo assessment. AAPS Pharmscitech. 2017;18:58–71.
  • Shreya AB, Managuli RS, Menon J, et al. Nano-transfersomal formulations for transdermal delivery of asenapine maleate: in vitro and in vivo performance evaluations. J Liposome Res. 2016;26:221–232.
  • Seidel EJ, Rother M, Regenspurger K, et al. A randomised trial comparing the efficacy and safety of topical ketoprofen in Transfersome(®) gel (IDEA-033) with oral ketoprofen and drug-free ultra-deformable Sequessome™ vesicles (TDT 064) for the treatment of muscle soreness following exercise. J Sports Sci. 2016;34:88–95.
  • Lu K, Xie S, Han S, et al. Preparation of a nano emodin transfersome and study on its anti-obesity mechanism in adipose tissue of diet-induced obese rats. J Transl Med. 2014;12:72.
  • Sarwa KK, Mazumder B, Rudrapal M, et al. Potential of capsaicin-loaded transfersomes in arthritic rats. Drug Deliv. 2015;22:638–646.
  • Ghanbarzadeh S, Arami S. Enhanced transdermal delivery of diclofenac sodium via conventional liposomes, ethosomes, and transfersomes. Biomed Res Int. 2013;2013:1–7.
  • Ghannoum M, Isham N, Henry W, et al. Evaluation of the morphological effects of TDT 067 (terbinafine in Transfersome) and conventional terbinafine on dermatophyte hyphae in vitro and in vivo. Antimicrob Agents Chemother. 2012;56:2530–2534.
  • Zhang YT, Xu YM, Zhang SJ, et al. In vivo microdialysis for the evaluation of transfersomes as a novel transdermal delivery vehicle for cinnamic acid. Drug. 2014;40:301–307.
  • Sigurgeirsson B, Ghannoum M. Therapeutic potential of TDT 067 (terbinafine in Transfersome): a carrier-based dosage form of terbinafine for onychomycosis. Expert Opin Investig Drugs. 2012;21:1549–1562.
  • Rajan R, Vasudevan DT. Effect of permeation enhancers on the penetration mechanism of transfersomal gel of ketoconazole. J Adv Pharm Technol Res. 2012;3:112–116.
  • Kaur CD, Saraf S. Topical vesicular formulations of Curcuma longa extract on recuperating the ultraviolet radiation-damaged skin. J Cosmet Dermatol. 2011;10:260–265.
  • Duangjit S, Opanasopit P, Rojanarata T, et al. Characterization and in vitro skin permeation of meloxicam-loaded liposomes versus transfersomes. J Drug Deliv. 2011;2011:418316.
  • Rother M, Seidel EJ, Clarkson PM, et al. Efficacy of epicutaneous Diractin (ketoprofen in Transfersome gel) for the treatment of pain related to eccentric muscle contractions. Drug Des Devel Ther. 2009;3:143–149.
  • Cevc G, Mazgareanu S, Rother M. Preclinical characterisation of NSAIDs in ultradeformable carriers or conventional topical gels. Int J Pharm. 2008;360:29–39.
  • Zheng Y, Hou SX, Chen T, et al. Preparation and characterization of transfersomes of three drugs in vitro. Zhongguo Zhong Yao Za Zhi. 2006;31:728–731.
  • Li S, Qiu Y, Zhang S, et al. Enhanced transdermal delivery of 18β-glycyrrhetic acid via elastic vesicles: in vitro and in vivo evaluation. Drug Dev Ind Pharm. 2012;38:855–865.
  • Chen G, Li D, Jin Y, et al. Deformable liposomes by reverse-phase evaporation method for an enhanced skin delivery of (+)-catechin. Drug Dev Ind Pharm. 2014;40:260–265.
  • Trotta M, Peira E, Debernardi F, et al. Elastic liposomes for skin delivery of dipotassium glycyrrhizinate. Int J Pharm. 2002;241:319–327.
  • Elsayed MM, Abdallah OY, Naggar VF, et al. Deformable liposomes and ethosomes: mechanism of enhanced skin delivery. Int J Pharm. 2006;322:60–66.
  • Cadena PG, Pereira MA, Cordeiro RBS, et al. Nanoencapsulation of quercetin and resveratrol into elastic liposomes. Biochim Biophys Acta. 2013;1828:309–316.
  • Gupta PN, Mishra V, Rawat A, et al. Non-invasive vaccine delivery in transfersomes, niosomes and liposomes: a comparative study. Int J Pharm. 2005;293:73–82.
  • Vanic Z, Hafner A, Bego M, et al. Characterization of various deformable liposomes with metronidazole. Drug Dev Ind Pharm. 2013;39:481–488.
  • Schmitt JV, Miot HA. Actinic keratosis: a clinical and epidemiological revision. An Bras Dermatol. 2012;87:425–434.
  • Memon AA, Tomenson JA, Bothwell J, et al. Prevalence of solar damage and actinic keratosis in a Merseyside population. Br J Dermatol. 2000;142:1154–1159.
  • Gupta AK, Paquet M, Villanueva E, et al. Interventions for actinic keratoses. Cochrane Database Syst Rev. 2012;12:CD004415.
  • Alvi IA, Madan J, Kaushik D, et al. Comparative study of transfersomes, liposomes, and niosomes for topical delivery of 5-fluorouracil to skin cancer cells: preparation, characterization, in-vitro release, and cytotoxicity analysis. Anticancer Drugs. 2011;22:774–782.
  • Khan MA, Pandit J, Sultana Y, et al. Novel carbopol-based transfersomal gel of 5-fluorouracil for skin cancer treatment: in vitro characterization and in vivo study. Drug Deliv. 2015;22:795–802.
  • Holmes SA, Malinovszky K, Roberts DL. Changing trends in non-melanoma skin cancer in South Wales, 1988-1998. Br J Dermatol. 2000;143:1224–1229.
  • Marks R, Staples M, Giles G. Trends in non-melanocytic skin cancer treated in Australia: the second national survey. Int J Cancer. 1993;53:585–590.
  • Miller DL, Weinstock MA. Nonmelanoma skin cancer in the United States: incidence. J Am Acad Dermatol. 1994;30:774–778.
  • Lear JT, Tan BB, Smith AP, et al. Risk factors for basal cell carcinoma in the UK: case-control study in 806 patients. J R Soc Med. 1997;90:371–374.
  • Corona R, Dogliotti E, Errico M, et al. Risk factors for basal cell carcinoma in a Mediterranean population. Arch Dermatol. 2002;137:1162–1168.
  • Gallagher RP, Hill GB, Bajdik CD, et al. Sunlight exposure, pigmentary factors, and risk of nonmelanocytic skin cancer. Arch Dermatol. 1995;131:157–163.
  • Yamada M, Kodama K, Fujita S, et al. Prevalence of skin neoplasms among the atomic bomb survivors. Radiat Res. 1996;146:223–226.
  • Maloney ME. Arsenic in dermatology. Dermatol Surg. 1996;22:301–304.
  • Hartevelt MM, Bavinck JN, Kootte AM, et al. Incidence of skin cancer after renal transplantation in the Netherlands. Transplantation. 1990;49:506–509.
  • Fadel M, Samy N, Nasr M, et al. Topical colloidal indocyanine green-mediated photodynamic therapy for treatment of basal cell carcinoma. Pharm Dev Technol. 2017;22(4): 545–550. ​
  • Friedman-Kien AE, Saltzman BR, Cao YZ, et al. Kaposi’s sarcoma in HIV-negative homosexual men. Lancet. 1990;335(8682):168–169.
  • Penn I. Kaposi’s sarcoma in organ transplant recipients: report of 20 cases. Transplantation. 1979;27:8–11.
  • Pathak K, Sharma V, Sharma M. Optimization, in vitro cytotoxicity and penetration capability of deformable nanovesicles of paclitaxel for dermal chemotherapy in Kaposi sarcoma. Artif Cells Nanomed Biotechnol. 2016;44:1671–1683.
  • Urba WJ, Washington CV, Nadiminti H. Cancer of the skin. In: Longo DL, Fauci AS, Kasper DL, editors. Harrison’s principles of internal medicine. 18th ed. New York, NY: McGraw-Hill Medical; 2012.
  • American Cancer Society. What are the key statistics about melanoma? [ cited 2017 Jan 01]. Available from: www.cancer.org/Cancer/SkinCancer-Melanoma/Detailed-Guide/melanoma-skin-cancer-key-statistics
  • O’Bryant CL, Poust JC. Melanoma. In: DiPiro JT, Talbert RL, Yee GC, editors. Pharmacotherapy: a pathophysiologic approach. 8th ed. New York: McGraw-Hill Medical; 2011.
  • Lin M-W, Huang Y-B, Chen C-L, et al. A formulation study of 5-aminolevulinic encapsulated in DPPC liposomes in melanoma treatment. Int J of Med Sci. 2016;13:483–489.
  • Dorrani M, Olga BG, Tamara M, et al. Development of edge-activated liposomes for siRNA delivery to human basal epidermis for melanoma therapy. J Control Release. 2016;228:150–158.
  • Marinkovich MP. Tumour microenvironment: laminin 332 in squamous-cell carcinoma. Nat Rev Cancer. 2007;7:370–380.
  • Lee DA, Miller SJ. Nonmelanoma skin cancer. Facial Plast Surg Clin North Am. 2009;17:309–324.
  • Akgul B, Cooke JC, Storey A. HPV-associated skin disease. J Pathol. 2006;208:165–175.
  • Moloney FJ, Comber H, O’Lorcain P, et al. A population-based study of skin cancer incidence and prevalence in renal transplant recipients. Br J Dermatol. 2006;154:498–504.
  • Morita A. Tobacco smoke causes premature skin aging. J Dermatol Sci. 2007;48:169–175.
  • Gupta V, Karthikeyan C, Trivedi P. Localized delivery of cisplatin for the effective management of squamous cell carcinoma from protransfersome formulation. Arch Pharm Res. 2012;35:51–859.
  • Ming K, Lin H, Juan W, et al. Enhanced transdermal lymphatic drug delivery of hyaluronic acid modified transfersomes for tumor metastasis therapy. J Royal Soc Chem. 2014;51(8):1453–1456.. ​
  • Paolino D, Licciardi M, Celia C, et al. Folate- targeted supramolecular vesicular aggregates as a new frontier for effective anticancer treatment in in vivo model. Euro J Pharm and Biopharm. 2012;82:94–102.
  • Alvi IA, Madan J, Kaushik D, et al. Comparative study of transfersomes, liposomes, and niosomes for topical delivery of 5-fluorouracil to skin cancer cells: preparation, characterization, in-vitro release, and cytotoxicity analysis. Anticancer Drug. 2011;22:774–782.
  • Mahmood S, Taher M, Mandal UK. Experimental design and optimization of raloxifene hydrochloride loaded nanotransfersomes for transdermal application. Int J Nanomedicine. 2014 12;9:4331–4346.
  • Bragagni M, Mennini N, Maestrelli F, et al. Comparative study of liposomes, transfersomes and ethosomes as carriers for improving topical delivery of celecoxib. Drug Deliv. 2012;19:354–361.
  • Maswadeh H, Demetzos C, Dimas K, et al. Accumulation of vinblastine into transfersomes and liposomes in response to a transmembrane ammonium sulfate gradient and their cytotoxic/cytostatic activity in vitro. Anticancer Res. 2001;21:2577–2583.
  • Cevc G. Preparation for the application of agents in mini-droplets. United States patent US 6165500 A. 2000.
  • Cevc G. Preparation for the transport of an active substance across barriers. United States patent US 20020048596 A1. 2002.
  • Perricone N. Topical drug delivery using phosphatidylcholine. United States patent US 20060105955 A1. 2006.
  • Cevc G. Formulation for topical non-invasive application in vivo. United States patent US 7175850 B2. 2007.
  • Cevc G. Preparation for the application of agents in mini-droplets. United States patent US 20070042030 A1. 2007.
  • Cevc G, Richardsen H, Weiland-Waibe A Method for the improvement of transport across adaptable semi-permeable barriers. United States patent US 7591949 B2. 2009.
  • William H, Henk-Andre K, Linda S. Methods of reducing the proliferation and viability of microbial agents. PCT WO2010/090654 A1. 2010.
  • Cevc G, Chopra A. Non-invasive vaccination through the skin. United States patent US 7867480 B1. 2011.
  • Richard WG, William H. Vesicles .PCT/European Patent. CA 2919971 A1. 2015.
  • Modi P. Stabilized and solubilized drug formulation for topical application and transdermal efficacy for cosmetic improvement and methods of formulation. United States patent US 20150157728 A1. 2015.