68
Universidade de Lisboa Faculdade de Ciências Departamento de Biologia Vegetal Development of nanoformulations of pyrazinoic acid prodrugs for treatment of Mycobacterium avium infections Mariana Cordeiro Matoso Mestrado em Microbiologia Aplicada 2012

Universidade de Lisboa - repositorio.ul.ptrepositorio.ul.pt/bitstream/10451/8460/1/ulfc102765_tm_mariana... · neste projecto bem como toda a disponibilidade concedida enquanto co-orientador

  • Upload
    ngohanh

  • View
    212

  • Download
    0

Embed Size (px)

Citation preview

Universidade de Lisboa

Faculdade de Ciências

Departamento de Biologia Vegetal

Development of nanoformulations of pyrazinoic

acid prodrugs for treatment of Mycobacterium

avium infections

Mariana Cordeiro Matoso

Mestrado em Microbiologia Aplicada

2012

Universidade de Lisboa

Faculdade de Ciências

Departamento de Biologia Vegetal

Development of nanoformulations of pyrazinoic

acid prodrugs for treatment of Mycobacterium

avium infections

Mariana Cordeiro Matoso

Dissertação orientada por Doutora Manuela Gaspar (FFUL) e

Prof. Doutora Sandra Chaves (FCUL)

Mestrado em Microbiologia Aplicada

2012

Development of nanoformulations of pyrazinoic

acid prodrugs for treatment of Mycobacterium

avium infections

Mariana Cordeiro Matoso

Master Thesis

2012

This thesis was fully performed at the Departamento de Farmácia

Galénica e Tecnologia Farmacêutica of the Faculty of Pharmacy

(University of Lisbon), Campus do Lumiar, under the direct

supervision of Doutora Manuela Gaspar and Prof. Doutor Luís

Constantino.

I

Acknowledgments

A realização desta Dissertação de Mestrado só foi possível graças à colaboração e ao

contributo, de forma directa ou indirecta, de várias pessoas e instituições, às quais gostaria de

exprimir algumas palavras de agradecimento e profundo reconhecimento, em particular:

À Doutora Manuela Gaspar pela disponibilidade manifestada para orientar este

trabalho. Agradeço todo o apoio, todas as horas perdidas de sono e todo o emaranhado de

pensamentos que tornaram possível a obtenção de boas soluções para alguns pequenos

contratempos que foram surgindo durante a realização deste trabalho e que, por certo,

causaram alguns cabelos brancos. Por todos os conhecimentos que me foram transmitidos e

que me ajudaram a tornar numa pequena adulta e por toda a amizade que foi sendo construída

durante este último ano, um muito obrigada.

Ao Prof. Doutor Luís Constantino pela oportunidade que me deu em poder trabalhar

neste projecto bem como toda a disponibilidade concedida enquanto co-orientador deste

trabalho. Por todas as discussões científicas que protagonizámos em conjunto com a Doutora

Manuela Gaspar e a Prof. Doutora Emília Valente e por todas as criticas que daí surgiram e

que me guiaram no bom caminho. Gostaria ainda de expressar o meu agradecimento pelo

fornecimento dos pró-fármacos utilizados durante esta dissertação.

Á Prof. Doutora Emília Valente agradeço a disponibilidade para as discussões

científicas realizadas e todas as críticas relativas ao trabalho.

À Doutora Eugénia Cruz, investigadora do grupo Nanomedicine & Drug Delivery

Systems da Faculdade de Farmácia da Universidade de Lisboa, pioneira na tecnologia de

lipossomas em Portugal, pela oportunidade concedida em poder trabalhar no laboratório do

Campus do Lumiar.

À Prof. Doutora Sandra Chaves por ter aceite ser minha orientadora da Faculdade de

Ciências, por toda a disponibilidade e pela revisão critica desta tese.

À Carla Eleutério pela paciência que teve em me educar e me ensinar a funcionar com

o HPLC. Por todo o carinho e amizade com que me recebeu e me aturou alguns devaneios e

crises existenciais.

À Alexandra Borges por ter cuidado tão bem dos “meus” ratinhos e por toda a alegria e

boa disposição que tornou os dias menos bons em dias excepcionais.

À Susana Calado e à Joana Pereira de Almeida pelo apoio e amizade que criámos e

que tão bem me fez à saúde e por todos aqueles momentos que às vezes parecem pequenos

mas que se tornam enormes.

II

Aos colegas e amigos Filipa Fontes e Rui Lopes por toda a amizade e apoio

demonstrados.

Às investigadoras e amigas Sandra Simões e Manuela Colla obrigada pela

disponibilidade e apoio prestados quando necessário.

Aos amigos de Sintra, Bruno, FT, Jises, Sandro, Sheila, Guida e Rute, por toda a

preocupação e pelas cafezadas tão boas que me permitiram desanuviar das semanas de

trabalho.

Ao meu namorado, João, um muito obrigada pelas longas horas de atenção

relativamente às minhas dúvidas, inquietações, desânimos e subvalorizações. Por todo o

apoio, confiança, valorização do meu trabalho e da minha pessoa e por toda a coragem que

me deu para ultrapassar a culpa pelo tempo que a cada dia lhe subtraía.

Aos meus pais, Lina e João, e à minha mana Madalena, por todos os conselhos e

incentivos que me deram e que me permitiram fazer mais e melhor nunca desistindo perante os

vários obstáculos e muros que se colocaram à minha frente. Obrigada!

Igualmente expresso o meu agradecimento à Fundação para a Ciência e Tecnologia

(FCT) que através do projecto A new life for old antimycobacterial drugs: development of

prodrugs of pyrazinoic acid activated by mycobacterial esterases as a way to circumvent

resistance to pyrazinamide, PTDC/SAU-FCF/101950/2008, contribuiu para o suporte financeiro

da presente dissertação.

III

Communications in Scientific Meetings

Stable Prodrugs Liposomes for Treatment of Mycobacterial Infections, Matoso M., Eleutério C.,

Constantino L., Cruz M.E.M., Gaspar M.M. 3rd iMed.UL Post--Graduate Students Meeting,

Faculdade de Fármacia Universidade de Lisboa, Lisboa, Portugal, p60

IV

Resumo

As micobactérias são bactérias Gram+ cujo diverso número de espécies actualmente

descritas (cerca de 130) partilham as mesmas características no que diz respeito à espessura

da parede celular e à sua composição única que as torna uma forte barreira à permeabilidade

de antibióticos. Algumas destas micobactérias têm um papel preponderante como agentes

patogénicos em indivíduos imunocompetentes como acontece com Mycobacterium

tuberculosis, Mycobacterium leprae and Mycobacterium ulcerans, os agentes causadores da

tuberculose (TB), lepra e úlcera de Buruli, respectivamente. A grande maioria das outras

espécies de micobactérias está presente no ambiente como microrganismos saprófitas,

podendo alguns ser considerados patogénicos oportunistas e causadores de morte em

indivíduos imunocomprometidos. Deste grupo de micobactérias ambientais oportunistas faz

parte o complexo Mycobacterium avium (MAC), composto por M. avium, subespécie avium,

paratuberculosis, silvaticum, e M. intracellulare, responsável por uma grande percentagem de

doenças micobacterianas não tuberculosas (NTM). As infecções provocadas por este complexo

são adquiridas através do ambiente, especialmente pela água, solo e alimentos. Apesar da

infecção poder ocorrer por inalação a principal via de infecção, especialmente em indivíduos

imunocomprometidos, é o tracto gastrointestinal, já que um sistema imunitário

imunocompetente consegue controlar estas micobactérias.

Os problemas associados aos tratamentos contra M. avium e M. tuberculosis são

provocados, entre outras causas, pela incapacidade dos antibióticos atravessarem a parede

celular complexa das micobactérias, atingindo concentrações sub-terapêuticas no interior dos

macrófagos, local de proliferação das micobactérias. A necessidade de ultrapassar esta

desvantagem utilizando doses mais elevadas dá origem ao aparecimento de efeitos tóxicos.

Uma vez que os esquemas de tratamento destas infecções são muito longos a adesão á

terapia é muitas vezes quebrada contribuindo para o desenvolvimento de resistências à grande

maioria dos antibióticos.

A pirazinamida (PZA) além de ser um antibiótico de primeira linha no tratamento da TB

é um pró-fármaco que requer activação pela enzima pirazinamidase (PZase) para ser

transformado em ácido pirazinóico (POA), a sua forma activa. Esta enzima é sintetizada pela

própria micobactéria que hidrolisa o pró-fármaco apenas dentro desta. No entanto, esta

activação está dependente apenas de uma enzima tendo sido observada a emergência de

estirpes resistentes à PZA. A origem destas resistências poderão ser devidas a mutações no

gene que codifica a PZase podendo não ocorrer a formação do POA uma vez que a enzima

não reconhece o seu substacto. De modo a ultrapassar estes problemas, têm sido investigadas

formas alternativas ao uso da PZA. Assim, a síntese de novos pró-fármacos que possam ser

activados por esterases micobacterianas e que, em virtude da sua elevada abundância,

possam contornar o desenvolvimento de resistências à PZA constitui uma excelente estratégia

terapêutica.

Nesta linha de investigação, novos pró-fármacos de POA foram investigados por

diversos autores tendo demonstrado actividade in vitro superior à PZA contra estirpes de M.

V

tuberculosis, M. avium e M. kansasii. No entanto, estes compostos apresentaram estabilidades

reduzidas na presença de fluidos biológicos. Recentemente, no grupo de Química Medicinal da

Faculdade de Farmácia da Universidade de Lisboa, foram sintetizados pró-fármacos de amidas

e ésteres de POA, com cadeias alcóxido de diferentes tamanhos. Os ésteres de POA

apresentaram elevada estabilidade em plasma, particularmente os de cadeia alcóxido longa.

Para além disso, estes pró-fármacos são facilmente activados pelas esterases

micobacterianas. Estudos in vitro demonstraram que estes ésteres são activos contra estirpes

sensíveis de M. tuberculosis em concentrações 10 vezes mais baixas do que as necessárias

para a PZA.

Na presente dissertação foram desenvolvidas formulações lipossomais de dois ésteres

de POA e o seu efeito terapêutico avaliado num modelo murino infectado com uma estirpe de

M. avium. A associação destes ésteres aos lipossomas é baseada no facto destes sistemas

lipídicos conseguirem contornar os problemas de solubilidade associados a estas moléculas e

ainda no facto de, após administração parentérica, poderem atingir o fígado e o baço, os

principais orgãos infectados por M. avium.

Na primeira parte desta tese foram desenvolvidas e caracterizadas formulações

lipossomais de dois ésteres de POA contendo 4 e 12 carbonos na cadeia alquílica linear,

designados por C4 e C12. Todas as formulações lipossomais de C12 apresentaram eficácias

de incorporação superiores a 90%. No entanto, para as formulações de C4 os valores obtidos

foram inferiores a 6% o que pode ser explicado pelo maior carácter lipofílico do C12 em

comparação com o C4.

Com base nestes resultados, o pró-fármaco C12 foi seleccionado para todos os

estudos efectuados a posteriori. Sendo assim, foram preparadas dois tipos de formulações

lipossomais: extrusadas e não extrusadas. Os resultados obtidos indicam que a razão final

entre o C12 incorporado e o lípido foi superior a 71 nmol/µmol de lípido para todas as

suspensões, independentemente do diâmetro e da composição lipídica, o que apoia a hipótese

do pró-fármaco estar incorporado na bicamada lipídica. Para fortalecer esta afirmação, foram

realizados estudos de calorimetria com lipossomas vazios e com C12 incorporado. Não se

observaram alterações nas temperaturas de transição de fase para as duas formulações. No

entanto, o aumento de entalpia registado na formulação lipossomal com C12 é o resultado da

interacção deste pró-fármaco com as cadeias lipídicas. Estas observações estão relacionadas

com o carácter lipofílico do C12, contribuindo para a rigidez do sistema lipossomal e

confirmando a sua localização na bicamada lipídica.

Na segunda parte deste trabalho foi realizado um estudo sistemático da estabilidade

das formulações de C12 na presença de tampão HEPES pH 7.4 e de plasma humano,

avaliando a velocidade de hidrólise de C12 e a respectiva formação de POA. Assim, a

quantificação destas duas moléculas foi realizada por HPLC, tendo-se procedido previamente à

optimização desta metodologia. A estabilidade em tampão foi realizada em duas condições

distintas: temperatura ambiente e 37ºC. A evolução do diâmetro das vesículas foi avaliada

durante um mês, para garantir a homogeneidade dos lipossomas aquando da sua utilização,

VI

não se verificando alterações nestas propriedades para as formulações em estudo. A

realização da estabilidade em tampão a 37ºC pretendeu avaliar se a percentagem de C12

incorporado se mantém ao longo de 24 horas. Os resultados obtidos demonstraram uma

elevada estabilidade de todos as suspensões relativamente ao pró-fármaco incorporado. Para

uma das formulações testadas (DMPC:DSPE-PEG) foi, ainda, analisada a concentração de

C12 e a possível formação de POA, tendo-se verificando que o pró-fármaco incorporado não

se encontrava na forma hidrolisada. A estabilidade das formulações na presença de plasma

humano foi também realizada a 37ºC a fim de avaliar a velocidade de hidrólise do pró-fármaco,

na forma livre e lipossomal, em função da composição lipidica, e de modo a estimar o seu

comportamento in vivo. Os resultados obtidos indicaram que a hidrólise do C12 foi influenciada

principalmente pelo diâmetro médio e rigidez das formulações testadas. De facto, os tempos de

semi-vida mais elevados foram obtidos para os lipossomas preparados com fosfolípidos

neutros e não submetidos a extrusão. A existência de um elevado número de bicamadas

lipídicas que caracterizam estas vesículas está na origem destes resultados. A incubação de

C12 na forma livre apresentou uma estabilidade superior comparativamente a algumas

formulações, particularmente para as vesículas extrusadas. No entanto, esta estabilidade foi

dependente da concentração inicial do pró-fármaco uma vez que, utilizando a dose terapêutica

usada no modelo animal infectado com M. avium testado neste trabalho, a velocidade de

hidrólise do C12 foi mais lenta nos lipossomas extrusados do que na forma livre.

Embora na literatura esteja descrito que as estirpes de M. avium são intrinsecamente

resistentes à PZA pretendeu-se avaliar se era possível com este pró-fármaco ultrapassar estas

desvantagens tendo em consideração os resultados promissores obtidos in vitro para os

ésteres de POA, contra a estirpe de M. tuberculosis. Foi a primeira vez que lipossomas deste

pró-fármaco foram testados num modelo murino infectado com M. avium. O estudo preliminar

permitiu demonstrar que após administração i.v. do inóculo os principais orgãos infectados são

o fígado e o baço. Tendo em consideração estes resultados foi avaliado o efeito terapêutico

das diferentes formulações em estudo através da contagem de unidades formadoras de

colónias (CFU) nestes dois orgãos. Paralelamente às formulações de C12 foi também avaliada

a actividade terapêutica de POA e PZA na forma livre. Os grupos de animais infectados e não

tratados evidenciaram valores de carga bacteriana mais elevados. O maior efeito terapêutico

foi observado para os grupos de animais tratados com C12 nas formas livre ou incorporado em

lipossomas extrusados, particulamente no fígado. Tendo em consideração que a estirpe de M.

avium utilizada neste modelo apresenta uma resistência superior à maioria dos antibióticos

comparativamente a algumas estirpes de M. tuberculosis os resultados obtidos são bastante

promissores.

Palavras-chave

Mycobacterium avium, pirazinamida, pró-fármacos, lipossomas, modelo animal

VII

Abstract

Pyrazinamide is a first line agent for tuberculosis treatment. Being a prodrug requires

activation by Mycobacterium enzyme pyrazinamidase to be converted into its active form,

pyrazinoic acid (POA). To overcome the emergence of resistant strains the synthesis of

prodrugs, esters of POA, which may be transformed in its active form, through mycobacterial

esterases were performed. Their high abundance contributes to a reduction in the emergence of

resistances.

In the present thesis, the development of liposomal formulations of two POA esters, with

different alkoxy chain lengths, and their therapeutic potential in an in vivo model infected

with Mycobacterium avium were performed. The association of POA esters to liposomes was

based on the ability of these lipidic systems to circumvent some solubility problems associated

to these molecules and, after parenteral administration, they can passively target liver and

spleen, the main affected organs in M. avium infections.

The incorporation parameters for these two prodrugs were dependent on their chain

length. Selecting the prodrug with higher incorporation efficiencies, in vitro and in vivo studies

were performed. A systematic study was performed in the presence of HEPES buffer pH 7.4

and biological fluids. In the buffer medium all liposomal formulations evidenced high stability in

terms of percentage of incorporated prodrug and preservation of non-hydrolyzed form. In

human plasma, the prodrug hydrolysis rate was mainly influenced by mean size and rigidity of

liposomal formulations: higher half-lives were obtained for neutral and non-extruded

liposomes. In vivo studies represent the first biological evaluation and comparison of new

synthesized esters of POA either in free or in liposomal forms. Extruded liposomes, although in

in vitro stability studies have shown a higher prodrug hydrolysis rate, leading to lower CFU/g of

liver than large sized vesicles. The use of other treatment schedules and mycobacterial strains

should be further considered.

Keywords

Mycobacterium avium, pyrazinamide, prodrugs, liposomes, animal model

VIII

List of Contents

Acknowledgments………………………………………………………………………………….…....I

Communications in Scientific Meetings……………………………………………………….......III

Resumo……………………………………………………………………………………………….....IV

Abstract……………………………………………………………………………………………........VII

List of Contents……………………………………………………………………………………….VIII

List of Figures…………………………………………………………………………………….……..X

List of Tables………………………………………………………………………………………...…XII

List of Abbreviations……………………………………………………………………..………….XIV

1. Introduction ................................................................................................................ - 1 -

1.1. Mycobacterial Infections........................................................................................ - 1 -

1.1.1. Nontuberculous Mycobacterial Diseases – The particular case of MAC ......... - 1 -

1.2. Conventional treatment of mycobacterial infections ............................................... - 2 -

1.3. Strategies to overcome antibiotic resistance .......................................................... - 3 -

1.3.1. Pyrazinamide ................................................................................................ - 3 -

1.3.2. Pyrazinoic Acid Prodrugs .............................................................................. - 4 -

1.3.3. Liposomes .................................................................................................... - 5 -

1.4. Mycobacterial murine models of infection .............................................................. - 6 -

1.5. Objectives of the thesis ......................................................................................... - 7 -

2. Materials and Methods ............................................................................................... - 8 -

2.1. Materials ............................................................................................................... - 8 -

2.1.1. Chemical products ........................................................................................ - 8 -

2.1.2. Animals ......................................................................................................... - 8 -

2.1.3. Mycobacterium avium strain .......................................................................... - 8 -

2.2. Methods ............................................................................................................... - 8 -

2.2.1. Preparation of prodrug liposomal formulations ............................................... - 8 -

2.2.1.1. Characterization of prodrug liposomal formulations .................................... - 9 -

2.2.1.1.1. Liposomal size measurements ......................................................... - 9 -

2.2.1.1.2. Zeta Potential Determination .......................................................... - 10 -

2.2.1.1.3. Prodrugs quantification .................................................................. - 10 -

2.2.1.1.4. Phospholipid quantification ............................................................. - 10 -

2.2.2. Differential scanning calorimetry (DSC) studies ........................................... - 11 -

2.2.3. HPLC System ............................................................................................. - 11 -

2.2.4. Stability of C12 formulations in presence of HEPES buffer........................... - 11 -

IX

2.2.5. Stability of C12 formulations in presence of human plasma ......................... - 12 -

2.2.5.1. Half-Life quantification of C12...................................................................... - 12 -

2.2.5.2. Stability of liposome structure in presence of human plasma ....................... - 12 -

2.2.6. Murine model .............................................................................................. - 13 -

2.2.6.1. Mycobacterium avium inocula quantification ................................................ - 13 -

2.2.6.2. In vivo evolution of mycobacterial infection .................................................. - 13 -

2.2.6.3. Biological evaluation of antimycobacterial formulations ................................ - 13 -

2.2.6.4. Evaluation of M. avium growth in mice ......................................................... - 14 -

2.3. Statistical analysis .............................................................................................. - 14 -

3. Results and discussion ............................................................................................ - 15 -

3.1. Physicochemical properties of esters of POA ...................................................... - 15 -

3.2. Incorporation of C12 and C4 in liposomes ........................................................... - 16 -

3.3. Optimization of HPLC procedures for quantification of C12 and POA .................. - 21 -

3.4. Stability of C12 formulations................................................................................ - 23 -

3.4.1. Stability of C12 formulations in HEPES buffer at room temperature ............. - 23 -

3.4.2. Stability of C12 formulations in HEPES buffer at 37ºC ................................. - 24 -

3.4.3. Stability of C12 formulations in human plasma at 37ºC ................................ - 25 -

3.4.3.1. Stability of C12 in the free form ................................................................ - 26 -

3.4.3.2. Stability of C12 liposomes – influence of lipid composition ....................... - 28 -

A) C12 incorporated in PC liposomes .............................................................. - 28 -

B) C12 incorporated in DMPC liposomes ......................................................... - 29 -

C) C12 incorporated in DMPC:DMPG liposomes .............................................. - 31 -

D) C12 incorporated in DPPC .......................................................................... - 32 -

E) C12 incorporated in DPPC:DPPG ............................................................... - 34 -

F) C12 incorporated in DMPC:DSPE-PEG ....................................................... - 35 -

3.4.3.3. Stability of C12 formulations – Influence of C12 concentration ................. - 37 -

3.4.3.4. Stability of liposome structure in presence of human plasma ................... - 39 -

3.5. Mycobacterium avium murine model of infection ................................................. - 40 -

3.5.1. Biological evaluation of antimycobacterial formulations ................................ - 40 -

3.5.2. In vivo evolution of mycobacterial infection .................................................. - 40 -

3.5.3. Influence of antimycobacterial formulations on M. avium murine model ....... - 41 -

4. Conclusions and Future Perspectives .................................................................... - 45 -

5. References ................................................................................................................ - 47 -

X

List of Figures

Figure 1. Action sites of the principal anti-TB drugs…………………………………………….……2

Figure 2. Zhang hypothesis for the mode of action of PZA……………………………………….…4

Figure 3. Cross section view of a liposome structure………………………………………………..5

Figure 4. Schematic representation of the experimental M. avium murine model of

infection…………………………………………………………………………………………….…….14

Figure 5. DSC thermograms. Influence of C12 on thermotropic behavior of DPPC

liposomes………………………………………………………………………………………………...20

Figure 6. Typical chromatogram of a C12 (a) and POA (b) freshly prepared solutions………...21

Figure 7. Calibration curves for C12 (a) and for POA (b)…………………….……………….……22

Figure 8. C12 hydrolysis over time…………………………………………………………………...23

Figure 9. POA formation over time due to C12 hydrolysis…………………………………………23

Figure 10. Stability on storage of C12 formulations: variation of mean size of C12 liposomes

during one month at room temperature……………………………………………………………….24

Figure 11. Stability in buffer, at 37ºC, of C12 formulations: influence of lipid

composition………………………………………………………………………………………..…….24

Figure 12. Concentration of C12 and POA after incubation in buffer at 37ºC..………………….25

Figure 13. Hydrolysis of C12 in free form and formation of POA in the presence of human

plasma 50% (v/v)………………………………………………………………………………….…….26

Figure 14. Hydrolysis of C12 in the free form in the presence of plasma for an initial

concentration of 60 µM…………………………………………………………………………………26

Figure 15. Hydrolysis of C12 incorporated in PC liposomes and formation of POA in the

presence of human plasma 50% (v/v)………………………………………………………………...29

XI

Figure 16. Hydrolysis of C12 incorporated in DMPC liposomes and formation of POA in the

presence of human plasma 50% (v/v)………………………………………………………………...30

Figure 17. Hydrolysis of C12 incorporated in DMPC:DMPG (7:3) liposomes and formation of

POA in the presence of human plasma 50% (v/v)…………………………………………………..31

Figure 18. Hydrolysis of C12 incorporated in DPPC liposomes and formation of POA in the

presence of human plasma 50% (v/v) ………………………………………………………….…….33

Figure 19. Hydrolysis of C12 incorporated in DPPC:DPPG (7:3) non-extruded liposomes and

formation of POA in the presence of human plasma 50% (v/v)………………………..…………..34

Figure 20. Hydrolysis of C12 incorporated in DMPC:DSPE-PEG (2.85:0.15) extruded liposomes

and formation of POA in the presence of human plasma 50% (v/v)…………………………. …..35

Figure 21. Half-life of C12 formulations after incubation in human plasma 50% (v/v)................36

Figure 22. Hydrolysis of C12 and formation of POA after incubation with human plasma 50%

(v/v)……………………………………………………………………………………………………….38

Figure 23. Absorbance at 420 nm of eluted C12 DMPC:DMPG extruded liposomes following

their application on the top of a Sephadex G200 column ……………………………………….…39

Figure 24. Evolution of CFU per g of organ in liver, spleen and lung of BALB/c mice 15 days

after infection induction ……………………………………………………………………………...…41

Figure 25. Evolution of M. avium infection in mice. ………………………...………………………42

Figure 26. . Evolution of M. avium infection in mice. Influence of administered formulations on

CFU per g of organ.…………………………………………………………………………………..…43

XII

List of Tables

Table 1. Structure and physicochemical properties of C4 and C12……………………………….15

Table 2. Physicochemical characterization of C12 and C4 extruded liposomes. Influence of

alkoxy chain length of prodrugs on incorporation parameters………………………...…………...17

Table 3. Physicochemical characterization of extruded and non-extruded C12 liposomal

formulations……………………………………………………………………………………………...19

Table 4. Detection and quantification limits for C12 and POA according to the respective

standards for each calibration curve…………………………………………………………………..22

Table 5. . kobs and half-life of free C12 in the presence of human plasma. Influence of initial

concentration…………………………………………………………………………………………….27

Table 6. kobs and half-life of free C12 in the presence of human plasma. Initial concentration: 60

µM……………………………………………………………………………………………………...…27

Table 7. kobs and half-life of C12 incorporated in PC liposomes in the presence of human

plasma: influence of mean vesicle size ………………………………………………………………29

Table 8. kobs and half-life of C12 incorporated in DMPC liposomes in the presence of human

plasma: influence of mean vesicle size …………………..…………………………………………30

Table 9. . kobs and half-life of C12 incorporated in DMPC:DMPG (7:3) liposomes in the presence

of human plasma: influence of mean vesicle size……………………….…………………………..32

Table 10. kobs and half-life of C12 incorporated in DPPC liposomes in the presence of human

plasma: influence of mean vesicle size……………………………………………………………….33

Table 11. kobs and half-life of C12 incorporated in DPPC:DPPG (7:3) non-extruded liposomes in

the presence of human plasma……………………………………………………………………..…34

Table 12. kobs and half-life of C12 incorporated in DMPC:DSPE-PEG (2.85:0.15) liposomes in

the presence of human plasma………………………………………………………………………..36

Table 13. kobs and half-life of C12 in free form and incorporated in DMPC:DMPG (7:3) extruded

liposomes in the presence of human plasma………………………………………………………...39

XIII

Table 14. Stability of liposome structure in the presence of human plasma - Mean size of

liposomes at time zero and 1 and 3h post-incubation following their application on the top of a

Sephadex G200 column………………………………………………………………………………..40

Table 15. C12 liposomal formulations used for the treatment of M. avium murine model of

infection.…………………………………………………………………………………………..……...41

XIV

List of Abbreviations

Ø Mean size

[(Cx/Lip)f Final Cx to Lipid ratio

[(Cx/Lip)i Inicial Cx to Lipid ratio

ΔH Enthalpy variation

ACN Acetonitrile

AIDS Acquired immune deficiency syndrome

ATCC American Type Culture Collection

CFU Colony forming units

D.L. Detection Limit

DMPC Dimyristoyl Phosphatidylcholine

DMPG Dimyristoyl Phosphatidylglycerol

DPPC Dipalmitoyl Phosphatidylcholine

DPPG Dipalmitoyl Phosphatidylglycerol

DSC Differential Scanning Calorimetry

DSMZ Deutsche Sammlung von Mikroorganismen und Zellkulturen

DSPE Distearoyl Phosphatidyl Ethanolamine

ETB Ethambutol

h hour

HIV Human immunodeficiency virus

HPLC High Performance Liquid Chromatography

I.E. Incorporation efficiency

INH Isoniazid

i.v. Intravenous

kobs First order rate constant

Lip Lipid

ln neperian logarithm

Ltd Limited

log Poct log octanol/water partition coefficient

XV

M. avium Mycobacterium avium

M. tuberculosis Mycobacterium tuberculosis

MAC Mycobacterium avium complex

min minute

MPS Mononuclear phagocytic system

NAD Nicotinamide adenine dinucleotide

non-MDR-TB non-Multi-Drug-Resistant-Tuberculosis

NTM Nontuberculous mycobacterial

OADC Oleic acid-albumin-dextrose-catalase

PC Egg Phosphatidylcholine

PdI Polydispersity index

PEG Polyethyleneglycol

POA Pyrazinoic Acid

PZA Pyrazinamide

PZase Pyrazinamidase

RIF Rifampicin

RT Retention time

s second

S.D Standard deviation

t1/2 Half-life time

Tc Phase transition temperature

TB Tuberculosis

UK United Kingdom

USA United States of America

- 1 -

1. Introduction

1.1. Mycobacterial Infections

Mycobacteria are Gram positive, nonspore-forming, aerobic bacteria. They are

considered facultative intracellular microorganisms presenting intracellular and extracellular

multiplication phases. Macrophages, resident phagocytes in different organs, are the main cells

that engulf invading microorganisms. Some pathogens that are internalized by macrophages

are transferred to lysosomal organelles followed by degradation. However, pathogenic

mycobacteria evade innate immunity by manipulating host, ensuring long-term survival and

proliferation that can occur in the phagosome (Gaspar et al., 2008a).

There are now over 130 known species of mycobacteria. However, only a few play an

important role as pathogenic agents for immunocompetent human hosts, namely

Mycobacterium tuberculosis, Mycobacterium leprae and Mycobacterium ulcerans, the causative

agents of tuberculosis (TB), leprosy and Buruli ulcer, respectively. The majority of other

Mycobacterium species are present in the environment as saprophytes, some of which can be

opportunistic and often deadly pathogens for immunocompromised human hosts. From this

group of environmental opportunistic mycobacteria, those from Mycobacterium avium complex

(MAC) are responsible for a large percentage of nontuberculous mycobacterial (NTM) diseases

(Reed et al., 2006).

1.1.1. Nontuberculous Mycobacterial Diseases – The particular case of

MAC

NTM diseases are acquired from environmental (water, soil) reservoirs and are not

transmitted between humans or between animals and humans. NTM infection progression to

clinical disease requires one or more predisposing host conditions. NTM species that cause

pulmonary disease vary by geographic region being MAC the predominant pathogen (Cook,

2010). In pre-AIDS (acquired immune deficiency syndrome) era, MAC, predominantly caused

pulmonary mycobacteriosis, that were difficult to distinguish radiologically from tuberculosis and

were rarely associated with extrapulmonary sites or disseminated diseases (Wolinsky, 1979).

Disseminated MAC became a frequent diagnosis in late-stage HIV (human immunodeficiency

virus) infected patients, usually occurring in patients with CD4+ T-cell counts below 50/mm

3.

Food-stuffs and water are the source of these organisms and the portal of entry in hosts is likely

to be the gastrointestinal tract. The syndrome is characterized by fever, shills, sweats, malaise,

weight loss, abdominal pain, and diarrhea (Cynamon and DeStefano, 1999). MAC refers to a

group of two closely related environmental mycobacteria: M.avium and M. intracellulare (Cosma

et al., 2003). Unlike TB, MAC disease is not required to be reported to public health entities in

most countries, and therefore, precise incidence and prevalence data are not available (Gaspar

et al., 2008a).

- 2 -

1.2. Conventional treatment of mycobacterial infections

Mycobacterial species share a characteristic thick cell wall, with a unique composition

that confers an exceptionally strong barrier to antibiotics, leading to resistances to a wide variety

of antimycobacterial agents. The lipid content of this cell wall gives rise to the common

characteristics of the Mycobacterium genus, which is the ability to retain basic dyes in presence

of an acid-alcohol solution (Cosma et al., 2003). The cell wall of mycobacteria is composed of

four layers: the first consists of peptidoglycan and the other three of a variety of soluble

proteins, carbohydrates, lipids and insoluble macromolecular components like arabinogalactan,

peptidoglycan and mycolic acid. The outer lipid barrier is extremely hydrophobic due to the

covalent bond between molecules of mycolic acid and arabinogalactan (Gaspar et al., 2008a).

The thick cell wall of mycobacterial species is in the origin of the high difficulty on the treatment

of mycobacterial infections. The most effective drugs to control M. tuberculosis infection are

Isoniazid (INH), Ethambutol (ETB), Pyrazinamide (PZA) and Rifampicin (RIF). These drugs are

first line therapeutic drugs and they represent the basis of tuberculosis therapeutic regiments. In

Figure 1 are shown their action sites in mycobacterial membrane: RIF inhibits bacterial RNA

synthesis by binding to the β-subunit of bacterial DNA-dependent RNA polymerase and

blocking the initiation chain formation in RNA synthesis; PZA is converted to Pyrazinoic Acid

(POA) and by decreasing the pH inside the mycobacterial cell prevents the microorganism

growth. PZA may also function as an antimetabolite of nicotinamide and interferes with the

synthesis of nicotinamide adenine dinucleotide (NAD), inhibiting the synthesis of short-chain,

fatty-acid precursors; ETB inhibits mycobacterial arabinosyl transferases involved in the

polymerization of D-arabinofuranose to arabinoglycan, an essential cell wall component; INH is

a prodrug activated by the mycobacterial catalase-peroxidase enzyme KatG inhibiting the

synthesis of mycolic acids, an essential component of mycobacterial cell walls (du Toit et al.,

2006).

Figure 1. Action sites of the principal anti-TB drugs. Adapted from du Toit et al., (2006)

Pyrazinamide

Isoniazid

Ethambutol

Rifampicin

- 3 -

In contrast with TB therapy, which is nearly uniformly effective when correctly administered

to non-Multi-Drug-Resistant-TB (non-MDR-TB), the therapy of MAC infections has historically

been less efficient and there are no standardized treatments (French et al., 1997; Georgiev,

1994; Nuermberger and Grosset, 2004). Therapeutic regimens include drugs such as

clarithromycin, RIF, Amikacin, Ciprofloxacin, ETB, Azithromycin, Rifabutin. However, an

increased number of mycobacterial strains have shown to be resistant to the available drugs. A

combinatory therapy for the treatment of these infections is recommended since monotherapy

gives rise to an enhanced occurrence of drug resistance and clinical failure.

1.3. Strategies to overcome antibiotic resistance

In an attempt to overlap the emergence of resistance strains to the available first-line

drugs, a viable strategy may be achieved by synthesis of new molecules or chemical

modification of already available ones. The design of new molecules such as prodrugs, entities

that need to undergo an enzymatic and/or chemical transformation in vivo to release the active

parent drug constitutes a good alternative. The development of prodrugs has become an

established tool for improving physicochemical, biopharmaceutical or pharmacokinetic

properties of pharmacologically active molecules and represent an increased tendency in

pharmaceutical industry (Rautio et al., 2008). In particular, the prodrug approach via activation

inside mycobacterial cells is a relevant alternative especially considering the problematic

penetration of drugs into these cells (Valente et al., 2011).

1.3.1. Pyrazinamide

PZA, a first line agent for the treatment of TB is also a prodrug. PZA plays an important

role in shortening TB therapy. This ability is related to its activity against a population of semi-

dormant mycobacteria residing in an acidic pH environment and that are not killed by other TB

drugs (Wade and Zhang, 2004; Zhang and Mitchison, 2003). In fact in vitro tests revealed a

lower activity in normal culture conditions close to neutral pH, while higher activity is observed in

acidic media (Wade and Zhang, 2004).

In order to have antimycobacterial activity, PZA requires activation by the

Mycobacterium enzyme pyrazinamidase (PZase) to be transformed into its active form, the

pyrazinoic acid (POA). This enzyme conveniently activates the drug only inside the bacteria

(Pires, 2011). The hypothesis for the mode of action of PZA according to Zhang and Mitchison,

(2003) is shown in Figure 2. PZA diffuses into the bacterial cell being then deaminated to form

POA. This molecule can only leave the cell again when it is excreted by an inefficient

mycobacterial efflux pump requiring energy (Mitchison and Fourie, 2010; Zhang et al., 2003).

- 4 -

Figure 2. Zhang hypothesis for the mode of action of PZA. Adapted from Mitchison and Fourie, (2010)

However, since the activation of PZA is dependent on only one enzyme, it has been

observed the emergence of resistant strains to this prodrug. As reviewed by Zhang and

Mitchison, (2003) the acquired PZA resistance in M. tuberculosis occurs by pncA gene mutation

(Raynaud et al., 1999).In other mycobacteria infections, in particular by M. avium, the natural

resistance to PZA may be due to an efficient POA efflux mechanism (Zhang and Mitchison,

2003). On the other hand the administration of POA, the active form of PZA, is not

recommended for treatment of mycobacteriosis due to its poor absorption and significantly

serum binding (Konno et al., 1967).

1.3.2. Pyrazinoic Acid Prodrugs

To overcome the already mentioned drawbacks, the synthesis of new POA prodrugs

were conducted by several authors (Cynamon et al., 1992; Cynamon et al., 1995; Fernandes et

al., 2010; Yamamoto et al., 1995). Cynamon et al., (1992) prepared series of POA esters and

evaluated their activity in vitro. Compared with PZA some compounds displayed higher activity

than PZA against M. tuberculosis, M. avium and M. kansasii (Cynamon et al., 1995). However,

these new compounds had lower stability in presence of biological fluids (Bergmann et al.,

1996).

Recently Simões et al. (2009) have synthesized series of pyrazinoic lipophilic prodrugs

esters with different alkoxy groups, and amide (Simões, 2005; Simões et al., 2009; Valente et

al., 2011). They observed that lipophilic esters with long alkoxy chains were more resistant to

plasma hydrolysis than the short chain esters (Simões et al., 2009). The selection of long chain

esters over amide prodrugs was based on the fact that lipophilic esters are easily activated by

mycobacterial esterases. This may contribute to a reduction in resistances to these prodrugs.

These esters were active in concentrations 10-fold lower than those needed for PZA to kill

sensitive M. tuberculosis in in vitro tests. On the other hand, it seems that amide prodrugs

cannot easily be activated by mycobacterial enzymes and thus have low in vitro activity (Simões

et al., 2009).

- 5 -

Besides the development of prodrugs with high in vitro activity, when applied to in vivo,

their pharmacological performance will be dependent on several factors, such as clearance or

metabolization after administration; distribution to infected sites, possible binding to plasmatic

proteins and the toxicity resulting from localization in non-affected sites, restricting the amount

of drug that can be administered (Gaspar et al., 2008a). A good strategy to overcome some of

these possible drawbacks could be the association of therapeutically promising molecules to

drug delivery systems. Ideally, these delivery systems should allow a preferential targeting of a

specific compound within a therapeutic concentration range at the affected sites; should be non-

toxic and non-immunogenic; should be biodegradable or easily excreted after exerting its effect;

and should be cheap and stable upon storage (Gaspar et al., 2008a). One of the most

extensively studied drug delivery systems are liposomes.

1.3.3. Liposomes

Liposomes, by definition, are submicron lipidic particles consisting of one or more

concentric lipid bilayers, separated by aqueous compartments (Gaspar et al., 2008a). It has

been more than 4 decades since the first report of the successful preparation of liposomes by

Bangham et al., (1965). There are several types of liposomes according to their lipid

compositions, number of lipid bilayers, superficial charges and mean sizes, ranging from few

nanometers to several microns. These liposomes can also be prepared using different methods

(Crommelin et al., 1994). Liposomes have been widely used for targeted drug delivery due to

their structural versatility, biodegradability, innocuous nature and resemblance to biological

membranes (Gregoriadis and Ryman, 1972). They can entrap drugs of different sizes and

solubility properties since the water-soluble compounds will be encapsulated in the aqueous

spaces while lipid-soluble ones will be incorporated in the lipid bilayer (Cruz et al., 2009;

Torchilin, 2005). In Figure 3 is shown the schematic structure of a liposome.

Figure 3. Cross section view of a liposome structure: (a) Hydrophilic head; (b) Hydrophobic tail; (c)

Internal aqueous space.

(Available at 22-08-2012 in http://www.britannica.com/EBchecked/media/92244/Phospholipids-can-be-

used-to-form-artificial-structures-called-liposomes )

Liposomes can be prepared according to the physicochemical properties of the molecule to

be incorporated and in order to reach the target either for therapy or for diagnosis (Cruz et al.,

2009). The structural and functional diversity of liposomes has been explored for the design of

- 6 -

drug carrier systems in particular for the treatment of mycobacterial infections (Fielding and

Lasic, 1999). After intravenous (i.v.) administration, some liposomes, depending on their

properties, are rapidly removed from blood circulation by the phagocytic cells of the

mononuclear phagocyte system (MPS), particularly by macrophages in liver and spleen. This

natural tendency has been exploited for treatment of infectious diseases localized in these

organs (Gaspar et al., 2008a). Intracellular infections caused by M. avium and M. tuberculosis

may be envisioned as a preferential target for liposomes The therapeutic advantages by

incorporating antibiotics in liposomes over the free respective molecules has been

demonstrated in several animal models of infection (Fielding and Lasic, 1999; Gaspar et al.,

2008a; Pinto-Alphandary et al., 2000). This superior therapeutic effect may be explained by the

different biodistribution profile of liposomal formulations in comparison with the free antibiotic

(Allen and Hansen, 1991; Allen and Stuart, 1999; Bakker-Woudenberg et al., 1993; Gaspar et

al., 2008b). However, the use of these type of liposomes, that are rapidly cleared from

bloodstream, may not be the right choice when other organs than liver and spleen are the

target. For inflammation and tumor pathological situations, that are not localized in liver and

spleen, the use of other type of liposomes with longer blood circulation half-lives represent

better solutions. Thus, the selection of lipid components is essential for the design of a

liposomal formulation. One successful hypothesis has been achieved by including in the lipid

composition the polyethyleneglycol (PEG), a polymer covalently linked to a phospholipid. The

presence of PEG at liposome surface is able to reduce plasmatic proteins adsorption and

consequently to decrease MPS clearance (Allen and Cullis, 2004; Torchilin, 2005). The

increased circulation time of liposomes has improved the delivery of therapeutic molecules to

infections localized in other organs than liver and spleen or even to inflammation and tumor

sites (Bakker-Woudenberg et al., 1993; Corvo et al., 2002; Gaspar et al., 1996; Gaspar et al.,

2007; Liu et al., 2006).

Therefore, the incorporation of esters of POA within liposomes appears as a good strategy

to enhance their in vivo antimycobacterial properties.

1.4. Mycobacterial murine models of infection

During the past years various animal models of MAC infection have been explored,

namely pigs, sheep, fowl, rabbits, goats, mice and guinea-pigs. However, the mouse is the most

widely used model of infection to evaluate the chemotherapeutic effect of antimycobacterial

drugs (Cynamon and DeStefano, 1999). Mice require small laboratory space, their purchase

and maintenance are relatively inexpensive and handling of these animals is easy if performed

by trained personal.

The induction of the infection may be perforrmed by a variety of routes including

intraperitoneal, intranasal, oral, intrarectal, aerogenic and i.v. The i.v. route is the most

commonly used as provides a consistent disseminated disease model with viable organisms

recovered from spleen, liver, blood, lung and lymph nodes. Usually for i.v. induction, mice are

infected in the tail vein. Each experiment consists of an early control group that is sacrificed at

- 7 -

the beginning of treatment and a later control that is sacrificed at the end of treatment schedule,

together with all treated groups. Primary cultures of mycobacteria species to be used for

infection may be obtained from clinical isolates or from bioresource centers such as American

Type Culture Collection, (ATCC) or from German Collection of Microorganisms and Cell Culture.

1.5. Objectives of the thesis

The main objectives in the present work were the development and characterization of

liposomal formulations of prodrugs of pyrazinoic acid (POA), particularly esters of POA, that in

previous in vitro tests have demonstrated to be active against M. tuberculosis (Simões et al.,

2009). The possible therapeutic effect of some of these liposomal formulations was tested in a

preliminary in vivo model infected with M. avium.

In order to fulfill the objectives of the present master thesis the work was executed

according to the following activities:

Development of best liposome methodologies for incorporating POA prodrugs in

liposomes. The influence of lipid composition, mean size, superficial charge and of the

physicochemical properties of POA prodrugs on incorporation parameters were studied.

Optimization of HPLC methodologies for evaluating the stability of the prodrug in buffer

and biological media. The comparison of chemical stability of the prodrug in free and

liposomal forms was performed.

Establishment of a M. avium model of infection that may be suitable for testing the

chemotherapeutic effect of POA prodrugs was carried out. Microbiological techniques

were used.

- 8 -

2. Materials and Methods

2.1. Materials

2.1.1. Chemical products

The prodrugs used C4 and C12 (esters of pyrazinoic acid) were previously synthesized

by the Medicinal Chemistry group of the Faculty of Pharmacy of the University of Lisbon

according to Simões et al., (2009). The following pure phospholipids were purchased from

Avanti Polar Lipids (USA): Dimyristoyl Phosphatidylcholine (DMPC), Dimyristoyl

Phosphatidylglycerol (DMPG), Dipalmitoyl Phosphatidylcholine (DPPC), Dipalmitoyl

Phosphatidylglycerol (DPPG), egg Phosphatidylcholine (PC) and Distearoyl Phosphatidyl

Ethanolamine covalently linked to Polyethyleneglycol (DSPE-PEG). Deionized water (Milli-Q

system; Millipore, Japan) was used for the preparation of all experimental solutions.

Middlebrook 7H9 broth and 7H10 agar, and Bacto Middlebrook albumin-dextrose-catalase and

oleic acid-albumin-dextrose-catalase (OADC) enrichments were obtained from Difco

Laboratories (USA). Nuclepore Track-Etch Membranes were purchased from Whatman Ltd,

USA. Kolliphor ELP from BASF, Germany, was kindly supplied by DS Produtos Químicos, Lda,

Portugal. All other reagents were of analytical grade.

2.1.2. Animals

Male BALB/c mice (5 to 7 weeks old, weight 25-30 g) were obtained from the

Gulbenkian Institute of Science (Portugal). The animals were kept under hygienic conditions,

fed commercial chow, and given acidified drinking water ad libitum. All the experimental

procedures were carried out with the permission of the local laboratory animal committee.

2.1.3. Mycobacterium avium strain

The bacterial strain used for the mouse model was M. avium 44157 from DSMZ

depository (Deutsche Sammlung von Mikroorganismen und Zellkulturen) and was kindly

provided by Unidade dos Retrovírus e Infecções Associadas of de Faculty of Pharmacy of the

University of Lisbon.

2.2. Methods

2.2.1. Preparation of prodrug liposomal formulations

Multilamellar vesicles composed of selected phospholipids were prepared as

previously described (Gaspar et al., 2000). The phospholipids and each prodrug used, in a

molar ration of 1:10, were previously solubilized in chloroform and dried by rotary evaporation

- 9 -

(Buchi, Switzerland) under nitrogen stream, to eliminate the organic solvent, until the formation

of a thin lipid film. The film was dispersed in deionized water, frozen and lyophilized (Edwards,

USA) overnight. Rehydration of lyophilized powder was made in two steps of 30 minutes (min)

each, at a temperature above the phase transition temperature (Tc) of the correspondent lipid

mixture. First rehydration was done in a volume of 2/10 of the final volume with 150 mM NaCl in

10 mM HEPES buffer, pH 7.4 (HEPES buffer, pH 7.4). After 30 min, rehydration was completed

with the same buffer. In order to reduce and homogenize the diameters of prodrug liposomes,

some formulations were submitted to an extrusion technique using an Extruder device (Lipex

Biomembranes Inc., Canada). Liposomal suspensions were sequentially filtered through

polycarbonate membranes of different porosities (0.8, 0.6, 0.4 and 0.2 µm) using a nitrogen

pressure ranging from 100 to 500 lb/in2 , until an average vesicle size between 0.1 – 0.2 µm was

achieved. The separation of non-incorporated prodrug from liposomes was performed by

ultracentrifugation (300 000 g for 2 hour (h) at 17ºC in a Beckman LM-80 ultracentrifuge

(Beckman Instruments, USA). The obtained pellet was ressuspended in the desired volume of

HEPES buffer, pH 7.4.

2.2.1.1. Characterization of prodrug liposomal formulations

Prodrug liposomal formulations were characterized in terms of lipid composition, lipid

and prodrug concentration, zeta potential, mean vesicle size and polydispersity index. The

following abbreviations and equations were used to determine these incorporation parameters:

(equation 1)

2.2.1.1.1. Liposomal size measurements

Liposome mean diameter was determined by dynamic light scattering in a

hydrodynamic sizing system (Zetasizer Nano S, Malvern Instruments, UK). This technique was

based on Brownian motion and was related to the size of the particles. For viscosity and

refractive index, the values of pure water were used. As a measure of particle size distribution

of the dispersion, the system reports the polydispersity index (PdI). The index ranges from 0.0

for an entirely monodisperse sample up to 1.0 for a polydisperse suspension. To determine the

- 10 -

mean diameter and PdI of liposomal preparations, a dilution to a final lipid concentration of 0.3

mM in HEPES buffer, pH 7.4 was previously performed, and samples were placed in an

appropriate polycarbonate cell. To ensure that appropriate mean diameter and PdI were

achieved, besides the measurements done for final liposomal preparations, these parameters

were also determined during the extrusion procedure.

2.2.1.1.2. Zeta Potential Determination

Zeta potential of liposomal formulations was measured in a hydrodynamic sizing system

(Zetasizer Nano Z, Malvern Instruments, UK). Zeta potential is defined as an electric potential

between the membrane surface and the ionic dispersion medium. This method determines how

fast a particle moves in a liquid when an electrical field is applied. For viscosity and refractive

index, the values of pure water were used.

Before determination of the zeta potential of liposomal formulations, an initial check of

the apparatus was made with a standard of a known zeta potential value (standard DTS5050,

Malvern Instruments, Ltd., UK). Dilutions of liposomal formulations were made in HEPES buffer,

pH 7.4 for a final lipid concentration of 0.3 mM. Samples were slowly introduced into a clear

disposable zeta cell with a syringe to avoid air bubbles. The zeta potential of samples at a

temperature of 25ºC was recorded. Three independent dilutions were prepared for each

liposomal formulation under study.

2.2.1.1.3. Prodrugs quantification

The prodrugs were quantified spectrophotometrically at 267 nm after disruption of the

liposomes with ethanol. Briefly, samples containing an amount of prodrug between 6.25 and

31.25 µM were pipetted in triplicate into 1.5 mL tubes and completed to 1 mL with ethanol. In

parallel, a calibration curve using a 2.5 mM stock prodrug solution and standards ranging from

6.25 to 31.25 µM were used. All tubes were shaken for 10 min and absorbances at 267 nm

were recorded in a UV 160 Spectrophotometer (Shimadzu, Japan).

The amount of prodrug in samples was obtained using a linear regression. The

calibration curve was linear up to absorbance values of at least 0.364.

2.2.1.1.4. Phospholipid quantification

The method for phospholipid quantification was based on the colorimetric determination

of PO43-

according to Rouser et al., (1970). Briefly, samples in triplicate containing a phosphate

amount between 20 and 80 nmol (sample volume less than 100 µL) were pipetted into 15 mL

glass tubes. In parallel, a calibration curve with phosphate amounts ranging from 20 to 80 nmol

was prepared using a 0.5 mM phosphate stock solution. All samples were then heated (180ºC)

in a heating block until their dryness. After cooling, 0.3 mL of perchloric acid (70-72%) was

added to all tubes. Marbles were placed on the top of all tubes and heated at 180ºC for 1 h to

convert all the organic lipid phosphate to inorganic form. After cooling samples to room

- 11 -

temperature, 1 mL of distilled water, 0.4 mL of hexa ammonium heptamolybate solution [1.25%

(w/v)], followed by 0.4 mL of ascorbic acid solution [5% (w/v)] were added to all glass tubes. All

tubes were heated in a boiling water bath for 5 min. During heating, the inorganic phosphate is

converted to phosphomolybdic acid due to reduction of ascorbic acid and a blue color is

developed. After cooling, the absorbance at 797 nm of all samples was recorded in a UV 160

Spectrophotometer (Shimadzu, Japan) against the blank of the calibration curve. The amount of

phosphate in samples was obtained using a linear regression. The calibration curve was linear

up to absorbance values of at least 1.000.

2.2.2. Differential scanning calorimetry (DSC) studies

The phase transition behavior of POA esters in liposomal formulations was performed in

a calorimeter DSC Q200 (TA Instruments, USA). Approximately 10 µL of liposomal

formulations (lipid concentration ca. 60 mM) were accurately measured into aluminum pans

which were hermetically sealed and then measured against an empty reference pan. The pans

were heated and the thermograms were recorded at a temperature ranging from 15 to 65ºC at

a heating rate of 3ºC/min.

2.2.3. HPLC System

All stability studies were made using C12. The stability of C12 was evaluated by HPLC

following an optimization of best conditions for quantifying the prodrug C12 and the

corresponding hydrolysis product, the POA, according to Simoes et al., (2009). The Beckman

System Gold HPLC system consisted of a 126 Pump Direct Control, the model detector 166

from Beckman Instruments, Inc and the Midas Spark 1.1 autoinjector. The wavelenght of the

detector was set at 267 nm. The analytical column was a LiChroCart® 125-4 Purospher Star

RP-8 (5 µm) (Merck, Germany). The system was attached to a computer with the software,

Katarat 7.7 for integration and treatment of chromatograms. The mobile phase, in an isocratic

solvent system, consisted in 75% of acetonitrile (ACN) for C12 and in 2% for POA in

KH2PO4/H3PO4 25 mM, (phosphate buffer, pH 2.0) with a flow rate of 1 mL/min. Calibration

curves for each compound, C12 and POA, were constructed with different standards ranging

from 5 to 25 µM. The limits of detection and quantification were calculated based on the

different standards used. All the quantifications were performed using calibration curves

constructed freshly prepared C12 and POA solutions.

2.2.4. Stability of C12 formulations in presence of HEPES buffer

The stability of C12 liposomes in HEPES buffer was evaluated by incubating the

formulations at 37ºC. At defined times, aliquots of suspensions were taken and applied onto the

top of a PD10 column to separate non incorporated C12. The fraction correspondent to

liposomes was analyzed for C12 and lipid contents. The mean size and superficial charge were

also analyzed along the incubation period. Different lipid compositions were characterized

(DMPC, DMPC:DSPE-PEG (2.85:0.15), DMPC:DMPG (7:3)). The stability was defined as the

- 12 -

ratio in percentage between C12 to lipid ratio at each studied time and the C12 to lipid ration at

time zero as shown in the following equation:

(equation 2)

,where tx are the ratios obtained at 0.5, 1, 4 and 24 h after incubation and t0 is the ratio at time

zero.

2.2.5. Stability of C12 formulations in presence of human plasma

The stability in plasma of C12 in free and liposomal forms was evaluated by incubating

the formulations in the presence of plasma diluted with HEPES buffer, pH 7.4 (50% v/v). The

hydrolysis of C12 and the formation of POA were determined by HPLC. At defined times 100 µL

of the reaction medium were 10 times diluted in ACN or in phosphate buffer, pH 2.0,

respectively for C12 and POA. Samples were centrifuged (Bench Centrifuge 202 MK, Sigma,

Germany) for 15 min, at 13 000 g at 17ºC, and the supernatant was filtrated with a 0.2 µm filter

and injected onto the HPLC system.

2.2.5.1. Half-Life quantification of C12

Apparent pseudo first-order kinetics and rate constants were determined, according to

Gupta et al., (2009), by using initial rates of hydrolysis. The apparent pseudo first-order

hydrolysis rate constants of C12 at 37ºC were determined by plotting the logarithm of C12

concentration as a function of time according to the equation:

(equation 3)

The hydrolysis half-live (in min) was then calculated by the equation:

(equation 4)

2.2.5.2. Stability of liposome structure in presence of human plasma

The evaluation of liposome structure in presence of human plasma was performed by

incubating liposomal formulations at 37ºC. At defined times aliquots of the suspension were

taken and applied onto the top of a Sephadex G200 column to separate liposomes from human

plasma. The fraction correspondent to liposomes was analyzed in terms of turbidity and

recorded by spectrophotometry at 420 nm. The mean size was also analyzed along the

- 13 -

incubation period. A single lipid composition was tested – DMPC:DMPG (7:3) extruded

liposomes.

2.2.6. Murine model

2.2.6.1. Mycobacterium avium inocula quantification

Inocula were prepared as previously described (Silva et al., 1987). Briefly, transparent

colonies of M. avium DSMZ 44157 were subcultured in Middlebrook 7H9 broth with albumin-

dextrose-catalase supplement and 0.04% Tween 80 (w/v) and allowed to grow at 37ºC on an

orbital shaker for 2 weeks. The bacteria were harvested by centrifugation (2 000 g, 10 min) in a

GPR Beckman centrifuge (Beckman Instruments, USA), suspended in a small volume of saline

with 0.04% Tween 80 (w/v), sonicated at low energy for 90 second (s) in a bath-type sonicator

(Bandelin Sonorex RK156, Germany) to disrupt bacterial clumps, diluted in the same medium to

an optical density of 0.8 at 600 nm and stored frozen at -70ºC until use. When needed, aliquots

were diluted to the desired concentration, and inoculated.

2.2.6.2. In vivo evolution of mycobacterial infection

BALB/c mice were injected intravenously with 5x105 colony forming units (CFU) of M.

avium DSMZ 44157. Fifteen days after infection, mice were sacrificed and their livers, spleens

and lungs were aseptically removed and homogenized. Homogenates were suspended and

serially diluted in 0.04% Tween 80 (w/v) and plated onto Middlebrook 7H10 agar medium

enriched with OADC for CFU counting after incubation at 37ºC for 10 to 15 days (Gaspar et al.,

2000).

2.2.6.3. Biological evaluation of antimycobacterial formulations

A murine model of M.avium infection previously described (Pedrosa et al., 1994) was

used. Each animal was infected by i.v. injection in a lateral tail vein with 1x106 CFU per mouse

in 200 µL of a M. avium suspension.

The therapeutic treatment started 2 weeks after the infection. On each week, treated

animals received three i.v. injections in a lateral tail vein during 2 weeks. The formulations under

study were C12 in the free form (solubilized in HEPES buffer pH 7.4 and Kolliphor ELP 5%

(w/v)) or incorporated in DMPC:DMPG (7:3) extruded and non-extruded liposomes. POA and

PZA previously solubilized in HEPES buffer pH 7.4 were also tested. The administered dose

was 12mg/kg of body weight related to POA.

- 14 -

2.2.6.4. Evaluation of M. avium growth in mice

Two days after the last treatment mice were killed by cervical dislocation and livers and

spleens were removed according to the method described in 2.2.6.2.

In Figure 4 a schematic representation of M. avium murine model of infection used in

this work is shown.

Figure 4. Schematic representation of the experimental M. avium murine model of infection

2.3. Statistical analysis

Data presented are expressed as mean (±) and standard deviation (S.D.). Statistical

analysis was performed using Single Factor ANOVA. The acceptable probability for a significant

difference between mean values was p<0.05 (Zar, 2009).

- 15 -

3. Results and discussion

3.1. Physicochemical properties of esters of POA

In previous work, developed by Simões et al. (2009) series of esters of POA, with a linear

alkoxy chain, ranging from 4 to 16 carbon atoms have been synthesized and stability studies performed

either in plasma or in macrophage cell cultures have demonstrated to be good candidates as

antituberculous drugs. Two of those compounds were selected for the present work to be incorporated

in liposomes: the esters containing 4 and 12 carbons linear chain length (C4 and C12, respectively).

The rational for associating these two esters of POA to liposomes was based on the fact that these lipid

systems are able to circumvent some solubility problems associated to these molecules. As widely

described in literature after parenteral administration liposomes can passively target their incorporated

material to infected cells in liver and spleen as is the case of M. avium infections (Cruz et al., 2009;

Gaspar et al., 2008a).

C12 and C4 are two prodrugs with different properties, which may influence their incorporation

in liposomes as well as their retention. In Table 1 are shown some physicochemical properties of these

two low molecular weight prodrugs, in particular the log octanol/water partition coefficient (log Poct.). The

increase on the carbon linear chain resulted on a concomitant enhancement on the log Poct from 0.9 to

4.55 for C4 and C12, respectively. The log Poct is a measure of the equilibrium concentration of a solute

between two immiscible phases: octanol and water. It may predict the potential for partitioning into

hydrophobic compartments such as lipid bilayers and hydrophilic compartments such as the aqueous

core of liposomes (Ribeiro et al., 2010). Generally, we can say that molecules with a log Poct higher than

1 present a lipophilic character while solutes with a log Poct below 1 are considered to be hydrophilic.

Moreover, log Poct > 5 correspond to high lipophilic compounds (Allen and Stuart, 1999; Defrise-

Quertain et al., 1984).

Table 1. Structure and physicochemical properties of C4 and C12

Data from Simões et al., (2009)

Prodrug R log Poct Molecular Weight

(MW)

C4 n-Butyl - (CH2)3CH3 0.9 180.2

C12 n-Dodecyl - (CH2)11CH3 4.55 292.4

- 16 -

3.2. Incorporation of C12 and C4 in liposomes

Due to the hydrophobic properties of C12 and C4, the liposome method for incorporating these

molecules in liposomes was the lipid film method, where both phospholipids and prodrugs were

solubilized in chloroform, submitted to an evaporation step, rehydrated with water, lyophilized overnight

and then rehydrated with a suitable buffer. This methodology has been widely used for incorporating low

molecular weight molecules of hydrophobic properties (Carvalheiro et al., 2009; Constantino et al.,

1993; Gaspar et al., 2000)

As described in materials and methods, two types of liposomes were used in the present work:

extruded and non-extruded. This technique constitutes one of the best methodologies to reduce and

homogenize the mean size of liposomes as it is suitable for the preparation of lipid systems in a scale

ranging from one to hundreds of mL (Cruz et al., 2009).

The influence of different lipid compositions on incorporation parameters was studied. Taking

this into consideration, the influence of phase transition temperature (Tc)* of the phospholipids as well

as the presence of negatively charged phospholipids in the lipid composition on the incorporation

efficiencies (I.E.) was compared for both prodrugs. The phospholipids tested were Dimiristoyl

Phosphatidylcholine (DMPC), Dimiristoyl Phosphatidylglycerol (DMPG) with a Tc of +23ºC and

Dipalmitoyl Phosphatidylcholine (DPPC) and Dipalmitoyl Phosphatidylglycerol (DPPG) with a Tc of

+41ºC. Liposomal formulations of C4 and C12 were prepared with neutral phospholipids (DMPC or

DPPC) and lipid mixtures containing also negatively charged phospholipids (DMPG or DPPG). In

addition, zeta potential, mean vesicle sizes and PdI were determined for all liposomal formulations.

In Table 2 are shown the physicochemical properties of extruded C12 and C4 liposomal

formulations.

*Tc values for the phospholipids referred above were obtained from Gunstone et al., (1986)

17

Table 2. Physicochemical characterization of C12 and C4 extruded liposomes. Influence of alkoxy chain length of prodrugs on incorporation parameters

Formulation nº

Lipid

Composition

(Molar Ratio)

Prodrug (Cx/Lip)i

(nmol/µmol)

(Cx/Lip)f

(nmol/µmol) I.E. (%)

Ø (µm)

(PdI)

Zeta Potential

(mV)

1 DMPC C12 117 ± 24 107 ± 16 92 ± 6 0.19

(0.4 - 0.5) -4 ± 1

2 DMPC C4 107 ± 1 8 ± 2 5 ± 2 0.18

(<0.2) -3 ± 1

3 DPPC C12 135 ± 11 103 ± 18 96 ± 10 1.3

(0.4 - 0.5) -2 ± 1

4 DPPC C4 129 ± 10 5 ± 1 4 ± 1 1.2

(0.5 - 0.6) -2 ± 2

5 DMPC:DMPG (7:3) C12 93 ± 5 85 ± 5 91 ± 4 0.16

(<0.2) -41 ± 4

6 DMPC:DMPG (7:3) C4 153 ± 4 7 ± 1 5 ± 1 0.16

(<0.2) -42 ± 4

7 DPPC:DPPG (7:3) C12 87 ± 2 95 ± 2 110 ± 6 0.17

(<0.2) -35 ± 3

8 DPPC:DPPG (7:3) C4 141 ± 3 6 ± 1 5 ± 1 0.19

(0.2 - 0.3) -39 ± 3

Initial lipid concentration – 20 mM; Initial prodrug concentration – 2 mM; [(Cx/Lip)f – Final Cx to Lipid ratio; [(Cx/Lip)i – Inicial Cx to Lipid ratio; Incorporation Efficiency I.E. (%) - [(Cx/Lip)f / (Cx/Lip)i)] * 100; Ø – mean size of liposomes; PdI – Polydispersity index.. Values correspond to mean ± S.D. of at least three independent preparations

18

C12 and C4 liposomal formulations presented a mean size below 0.2 µm with the

exception of liposomes prepared with the phospholipid DPPC (Formulations 3 and 4). For these

two formulations the mean vesicle sizes were higher than 1.2 µm after final preparation of

liposomes even using the extrusion step. DPPC is a neutral and rigid phospholipid with a Tc of

+41ºC and the possibility of aggregation due to its low superfical charge may be one of the

reasons of these high mean diameters (Crommelin and Schreier, 1994). For the other neutral

formulations (Formulations 1 and 2), prepared with a less rigid phospholipid (DMPC), this

aggregation effect was not observed. The zeta potential observed for Formulations 1, 2, 3 and 4

ranged from -2 to -4 mV. C12 and C4 liposomes containing in the lipid composition the

negatively charged phospholipids DMPG or DPPG, gave rise to formulations with a superficial

charge in agreement with their constituints: zeta potential values ranged from -35 to -41 mV. In

terms of loading capacity, a direct influence on the length chain of POA esters was observed.

While for C12 liposomal formulations an I.E. higher than 90% was achieved for all tested lipid

compositions (neutral and negatively charged) independently from Tc of the phospholipids used,

C4 formulations presented I.E. below 6% irrespectively from the lipid mixture used. This can be

explained by the less lipophilic character of C4 in comparison to C12. As above presented in

Table 1, a high difference on log Poct for these two prodrugs was observed: 0.9 and 4.55 for C4

and C12, respectively. The most favorable conditions for the maintenance of a solute within

liposomes correspond to extremely low or extremely high log Poct , while molecules with an

intermediate log Poct, such as C4, are poorly retained in liposomes (Allen and Stuart, 1999;

Defrise-Quertain et al., 1984). Based on these results the following studies were only performed

with C12. In conditions where molecules are poorly retained in liposomes, other liposome

preparation methods may be used particularly in case of weak acids or weak bases. The

incorporation of these molecules may be carried out in pre-formed liposomes in response to a

pH or salt gradient due to the higher permeability of neutral over charged molecules through

lipid bilayers. This strategy has been successfully used for weak bases such as doxorubicin or

vincristine (Allen et al., 1995; Bolotin et al., 2012).

A physicochemical comparison for extruded and non-extruded liposomes was

performed for C12 and the obtained results are shown in Table 3. The main interest was to

evaluate the influence of extruded and non-extruded liposomes on hydrolysis rate of this

prodrug in presence of human plasma. These studies were a first screening and that is the

reason why some formulations were not prepared in triplicate.

Besides the lipid compositions already tested and shown in Table 2, C12 was also

incorporated in liposomes prepared with a more fluid phospholipid, PC (Tc of -6ºC) and with a

lipid mixture of DMPC with DSPE-PEG. In addition, for the lipid mixture DMPC:DMPG three

different formulations (Formulation 5, 13 and 14) in terms of mean size were prepared and the

respective incorporation parameters compared.

19

Table 3. Physicochemical characterization of extruded and non-extruded C12 liposomal formulations

Formulation nº

Lipid

Composition

(Molar Ratio)

Extruded / Non-

Extruded

(C12/Lip)i

(nmol/µmol)

(C12/Lip)f

(nmol/µmol) I.E. (%)

Ø (µm)

(PdI)

Zeta Potential

(mV)

9 PC Non-Extruded 93 72 78 3.3

(0.7 - 0.8) -2 ± 1

10 PC Extruded 116 74 64 0.2

(<0.2) -2 ± 1

11 DMPC Non-Extruded 89 ± 6 96 ± 13 108± 7 6.5

(0.3 - 0.4) -3 ± 1

1 DMPC Extruded 117 ± 24 107 ± 16 92 ± 6 0.19

(0.4 - 0.5) -4 ± 1

12 DPPC Non-Extruded 68 ± 1 64 ± 5 94 ± 6 3.8

(0.8 - 0.9) -2 ± 1

3 DPPC Extruded 135 ± 11 103 ± 18 96 ± 10 1.3

(0.4 - 0.5) -2 ± 1

13 DMPC:DMPG (7:3) Non-Extruded 81 ± 10 71 ± 11 86 ± 11 1.7

(0.9 - 1) -33 ± 2

14 DMPC:DMPG (7:3) Extruded 73 70 96 0.55

(0.4 - 0.5) -35 ± 1

5 DMPC:DMPG (7:3) Extruded 93 ± 5 85 ± 5 91 ± 4 0.16

(<0.2) -41 ± 4

15 DPPC:DPPG (7:3) Non-Extruded 72 ± 3 78 ± 12 101 ± 10 0.96

(0.4 - 0.5) -33 ± 1

7 DPPC:DPPG (7:3) Extruded 87 ± 2 95 ± 2 110 ± 6 0.17

(<0.2) -35 ± 3

16 DMPG:DSPE-PEG

(2.85:0.15) Extruded 93 89 95

0.15

(<0.2) -5 ± 1

Initial lipid concentration – 20 mM; Initial C12 concentration – 2 mM [(C12/Lip)f – Final C12 to Lipid ratio; [(C12/Lip)i – Inicial C12 to Lipid ratio; Incorporation Efficiency I.E. (%) - [(C12/Lip)f / (C12/Lip)i)] * 100; Ø – mean size of liposomes; PdI – Polydispersity index. Values correspond to mean ± S.D. of at least three independent preparations when applicable

20

Although the initial conditions were the same for the preparation of all liposomes, the

experimental values obtained presented high variations (from 68 ± 1 to 135 ± 11). These may

be attributed to problems during weighing process of liposome constituents, loss of material

during lyophilization step or due to inexistence of triplicates. Nevertheless, the preparation of

extruded and non-extruded liposomes presented similar incorporation parameters for the same

lipid composition. Taking into account the formulations that were prepared in triplicate and,

consequently, presenting more reliable values, the final C12 to lipid ratio ranged from 71 ± 11 to

107 ± 16 either for extruded or non-extruded liposomes independently from lipid composition.

These results support the hypothesis that C12 was incorporated in the lipid bilayer.

To confirm the above results, calorimetric studies were preformed with unloaded and loaded

liposomes (liposomes without C12 and with C12 incorporated, respectively) to evaluate the

influence of C12 on thermal behavior of lipid bilayers. Differential Scanning Calorimetry (DSC)

has been extensively used to evaluate possible changes on conformational properties of

phospholipid membranes (Taylor and Morris, 1995). DPPC, the highest rigid phospholipid used

in these studies, was selected as even after submitting this formulation to extrusion procedures

it was not possible to obtain vesicles with a mean size below 0.2 µm. In Figure 5 are shown the

thermograms obtained with the respective Tc and enthalpies (ΔH).

Figure 5. DSC thermograms. Influence of C12 on thermotropic behavior of DPPC liposomes. Tc and ΔH for unloaded and loaded liposomes

According to obtained results, no differences were observed in terms of Tc for unloaded

and loaded liposomes. Both formulations presented a Tc of +41ºC; that corresponds to the

transition from a gel state to a liquid crystal state of the constituent phospholipid. However, an

increase on the enthalpy was observed when C12 was incorporated in the lipid bilayer. This

may be due to an interaction of the prodrug with the lipid chains that is related to its lipophilic

properties, leading to a higher rigidity of the lipid bilayer and confirming the localization of C12 in

the lipid matrix. These observations are in accordance with literature (Castelli et al., 2003;

Demetzos, 2008) and may explain the increase on the mean size of C12 DPPC liposomes,

even using an extrusion step, since the rigidity influences the mean size of the vesicles (see

Formulation 3 in Tables 2 and 3). These calorimetric studies were only performed with

21

liposomes prepared with DPPC. Nevertheless, we may expect that the same effect should be

observed using other phospholipids.

3.3. Optimization of HPLC procedures for quantification of C12 and

POA

The quantification of C12 for the physicochemical characterization of liposomes was

performed by spectrophotometric techniques.

The chemical stability studies of C12 formulations in the presence of buffer and human

plasma were intended to evaluate the hydrolysis rate of C12 and the respective formation of

POA. In order to perform these studies, the quantification of C12 and POA was performed by

HPLC. Thus, an optimization of this methodology was carried out as described by Simões et al.,

(2009). In Figure 6 is shown a typical chromatogram for C12 and for POA from freshly prepared

solutions.

Figure 6. Typical chromatogram of a C12 (a) and POA (b) freshly prepared solutions. The eluents consisted in 75% of ACN for C12 and in 2% for POA in phosphate buffer, pH 2.0 with a flow rate of 1 mL/min. C12 retention time (RT) – 5.24 min; POA RT – 4.31 min

At least three independent calibration curves were constructed for each compound. The

graphical representations of the different sets of calibration curves are shown in Figure 7 as well

as the respective values of the linear regressions.

22

(a) (b)

Figure 7. Calibration curves for C12 (a) and for POA (b). The results represent mean ± S.D. of all standard curves. The linear regression was calculated using the method of least squares

The obtained results evidence a high linearity and reproducibility in the selected ranges

from batch to batch as a variation below 10% was observed from experiment to experiment.

For both molecules, and regarding the respective calibration curves, it was determined

the detection and quantification limits according to Miller, (1991). Results are shown in Table 4.

Table 4. Detection and quantification limits for C12 and POA according to the respective standards for each calibration curve

Molecule Detection Limit (µM) Quantification Limit (µM)

C12 0.173 0.523

POA 1.069 3.241

The HPLC optimization allowed the evaluation of C12 stability and the formation of POA

over time. In Figure 8 a) is represented the chromatogram of a freshly prepared C12 solution.

The reduction of C12 and the formation of POA are shown in Figures 8 b), c) and 9 b), c),

respectively.

23

Figure 8. C12 hydrolysis over time: a) Freshly C12 solution; b) and c) C12 solution along hydrolysis process

Figure 9. POA formation over time due to C12 hydrolysis: a) Freshly C12 solution; b) and c) increase of POA formation due to C12 hydrolysis

3.4. Stability of C12 formulations

The stability of some C12 liposomal formulations above mentioned was evaluated

following their incubation in HEPES buffer and biological fluids. The stability in buffer was

analyzed using two different conditions: room temperature and 37ºC. The first one was

performed in order to analyze the stability of liposome mean size over time. The incubation at

37ºC had the purpose of evaluating if the properties of liposomal formulations, particularly the

percentage of C12 incorporated, were maintained even after being submitted to severe

conditions. These stability studies intent to select the formulations that are able to preserve their

properties in terms of mean size and incorporated prodrug for further evaluation in in vivo

animal models. On the other hand, the stability studies in biological fluids were performed in

order to evaluate the hydrolysis rate of the prodrug and thus to estimate its in vivo behavior.

3.4.1. Stability of C12 formulations in HEPES buffer at room temperature

The stability on storage at room temperature of C12 liposomes was evaluated during

one month by the determination of mean sizes, as described in materials and methods. The

selected lipid compositions in this study were DMPC, DPPC, DMPC:DMPG and DPPC:DPPG

and all of them were submitted to extrusion procedures. Figure 10 shows the obtained results.

24

0 10 20 300,0

0,1

0,2

1,0

1,2

1,4

1,6

1,8

2,0

Me

an

Siz

e (

µm

)

Time (days)

DMPC

DPPC

DPPC:DPPG

DMPC:DMPG

Figure 10. Stability on storage of C12 formulations: variation of mean size of C12 liposomes during one month at room temperature. Lipid compositions under study: DMPC, DPPC, DMPC:DMPG (7:3) and DPPC:DPPG (7:3)

In the beginning of the study all formulations presented a diameter below 0.2 µm with

the exception for DPPC liposomes, with a mean size around 1.6 µm, regardless of being

extruded.

During the analyzed period, all C12 liposomal formulations presented high stability as

no particular differences in terms of mean size were observed. The highest variation was

observed for the lipid composition DPPC but in the beginning of the experiment this formulation

was already very heterogeneous.

3.4.2. Stability of C12 formulations in HEPES buffer at 37ºC

The stability of C12 liposomes, in the presence of HEPES buffer, was evaluated

following their incubation at 37ºC. For this study C12 was incorporated in DMPC, DMPC:DSPE-

PEG and DMPC:DMPG (with two different mean sizes). At defined times aliquots of each

suspension were taken and applied onto the top of a PD10 column to separate non-

incorporated C12. The fraction correspondent to liposomes was determined in terms of C12 and

lipid contents. The influence of lipid composition and mean size, particularly for DMPC:DMPG

formulations, on the preservation of C12 liposomes properties was analyzed. The obtained

values for C12 to lipid ratio related to initial conditions expressed in percentage, according to

equation 2 in materials and methods, are represented in Figure 11.

Figure 11. Stability in buffer, at 37ºC of C12 liposomal formulations: influence of lipid composition. Lipid compositions tested: DMPC - 0.19 µm, DMPC:DSPE-PEG (2.85:0.15) - 0.15 µm, DMPC:DMPG A (7:3) - 0.16 µm, DMPC:DMPG B (7:3) - 0.55 µm

25

Taking into account these results, for all lipid compositions under study, a high stability

of C12 when incorporated in liposomes was demonstrated, as 24 h post-incubation more than

95% of C12 was still associated to these vesicles.

Mean size and zeta potential were also determined in the beginning and at the end of

incubation period for all formulations and no differences were observed (data not shown).

The quantification of C12 (Figure 11) was performed by spectrophotometry and so it

was not possible to evaluate if part of the prodrug was already hydrolyzed. Thus, a liposomal

formulation was chosen randomly (DMPC:DSPE-PEG) to conduct a more detailed study by

HPLC. The C12 concentration and the possible formation of POA were determined using

aliquots recovered from the PD10 column. These quantifications meant to evaluate if the

incubation of C12 liposomes, at 37ºC, was able to induce the hydrolysis of the prodrug. The

obtained results are shown in Figure 12.

Figure 12. Concentration of C12 and POA after incubation in buffer at 37ºC. Lipid Composition – DMPC:DSPE-PEG (2.85:0.15)

The amount of POA obtained by HPLC was below detection limit (D.L.) while a constant

value for C12 was achieved for all analyzed samples proving that, under these experimental

conditions, C12 incorporated in liposomes did not undergo hydrolysis.

Data obtained shows a high stability of C12 incorporated in liposomes for the lipid

composition DMPC:DSPE-PEG both in terms of percentage of incorporated prodrug (Figure 11)

and in the ability to preserve the non-hydrolyzed form (Figure 12).

3.4.3. Stability of C12 formulations in human plasma at 37ºC

The stability in plasma was performed, as already mentioned, to evaluate the hydrolysis

rate of the prodrug and the formation of POA and thus to estimate its in vivo behavior. The

obtained results were compared to the respective profiles after prodrug incorporation in

liposomes. In addition, the influence of lipid composition and mean size of liposomes on C12

hydrolysis rate was analyzed.

26

3.4.3.1. Stability of C12 in the free form

A stock solution of C12 was prepared and the hydrolysis rate of this prodrug and the

formation of POA were monitored using two different initial concentrations: 24 and 60 µM.

Figure 13 shows the hydrolysis of C12 and the consequent formation of POA for these two

experimental conditions.

(a) (b)

Figure 13. Hydrolysis of C12 in free form and formation of POA in the presence of human plasma 50% (v/v). (a) Initial concentration: 24 µM. (b) Initial concentration: 60 µM. The results represent mean ± S.D. of two independent experiments

According to obtained results, the reduction of C12 was always followed by a

concomitant increase on POA in both experimental conditions. Particularly, when using the

highest C12 initial concentration, at time zero, this value was approximately 50 µM and 180 min

post-incubation the amount of POA determined was very similar (46 µM).

The apparent first-order hydrolysis rate constants of C12 at 37ºC were determined by

plotting the ln of C12 concentration as a function of time according to equations 3 and 4 in

materials and methods. As an example, in Figure 14 is represented data obtained for hydrolysis

of C12 in free form. The same procedure was executed for all the other formulations studied in

the present work.

Figure 14. Hydrolysis of C12 in free form in the presence of plasma for an initial concentration of 60 µM. Evolution of ln of C12 concentration over time

27

The half-lives and the respective kobs values for C12 in free form for the two studied

concentrations are shown in Table 5.

Table 5. kobs and half-life of free C12 in the presence of human plasma. Influence of initial concentration

Initial concentration (µM) kobs x 103 (min

-1) Half-life time (min)

24 13.9 ± 0.2 50 ± 3

60 16.6 ± 1.6 43 ± 4

No statistically significant differences were observed on the half-lives obtained for the

two initial concentrations (p>0.05).

When this study was performed using the lower initial concentration the hydrolysis rate

of C12 was only evaluated until a reduction of 50% on initial amount was reached. However,

using a 60 µM C12 concentration, it was possible to determine its reduction to 50%, 75% and

87.5% related to the initial amount because the study was evaluated for a longer period of time.

Results are shown in Table 6.

Table 6. kobs and half-life of free C12 in the presence of human plasma. Initial concentration: 60 µM

Percentage of initial C12

concentration (%) kobs x 10

3 (min

-1) Half-life time (min)

50 16.7 ± 0.1 38 ± 3

25 18.9 ± 2.9 38 ± 6

12.5 16.6 ± 1.6 43 ± 4

Regardless of the reduction on the percentage of initial C12 concentration, ranging from

50 to 87.5%, no statistical significant differences were observed in terms of the respective half-

lives determined (p>0.05).

According to literature, the hydrolysis rate of prodrugs in presence of plasma, for

determination of the respective half-lives, should be evaluated until a reduction of 87.5% on the

initial amount of the molecule is achieved (Maskill, 1989). In the stability studies performed in

the present work, and for some formulations, the hydrolysis rate of the prodrug was not

observed long enough, and thus a reduction on the initial C12 concentration to 12.5% was not

achieved. Nevertheless, the main goal in this systematic study was the achievement of a

general comparison regarding the hydrolysis profile of the prodrug according to lipid

composition and mean size of liposomes.

In a previous published work, the half-life obtained for this ester of POA was lower, less

than 17 min (Simões et al., 2009). However, the experimental conditions were different as a

higher percentage of human plasma was used (80% (v/v)). In present studies the percentage of

28

human plasma was 50% (v/v) and consequently the possibility of C12 hydrolysis is also

diminished. Nevertheless, the stability studies in human plasma, in the present work, were

always performed using the same percentage of this biological fluid and thus the validity of the

comparisons made between the free and the liposomal forms remains. The hydrolysis rate of

C12 in the liposomal form, and the respective influence of different lipid compositions and

concentrations will be compared with the obtained C12 half-life presented in Table 6 due to the

superior number of analyzed samples.

3.4.3.2. Stability of C12 liposomes – influence of lipid composition

In a previous work (Simões, 2005) C12 was already incorporated in liposomes.

However the mean size of those vesicles was not determined and according to microscope

visualization those lipid structures presented very high and heterogeneous sizes (> 3 µm). In the

present work we intended to study the influence of mean size and lipid composition of C12

liposomes on hydrolysis rate. The reason for studying these two parameters is based on the

fact that mean size and lipid composition, among others, are able to influence liposomes in vivo

fate. With this purpose a systematic study was performed using several C12 liposomal

formulations prepared with phospholipids of different phase transition temperatures (Tc). For this

purpose phospholipids with increased Tc were used: A) PC, (Tc -6ºC); B) DMPC and D) DPPC.

In addition, for phospholipids B) and D) the presence of negatively charged phospholipids with

similar Tc (C) DMPG (Tc- +23ºC) and (E) DPPG (Tc - +41ºC) was also analyzed.

A) C12 incorporated in PC liposomes

C12 was incorporated in PC, a neutral and fluid phospholipid with Tc below 0ºC (-6ºC).

Two different liposome suspensions were prepared: non-extruded and extruded, with a mean

size of 3.3 µm and 0.2 µm, respectively. The stability of these two C12 formulations was

performed after incubation in human plasma and the hydrolysis of the prodrug and the formation

of POA were determined.

29

(a) (b)

Figure 15. Hydrolysis of C12 incorporated in PC liposomes and formation of POA in the presence of human plasma 50% (v/v). Initial concentration: 60 µM. (a) Non-extruded liposomes. (b) Extruded liposomes. The results represent mean ± S.D. of two independent experiments

According to obtained results a faster depletion of C12 and the corresponding formation

of POA was observed for extruded liposomes (Figure 15 (b)) in comparison with non-extruded

vesicles (Figure 15 (a)). Moreover, the hydrolysis profile for extruded liposomes presented a

more linear variation over time than non-extruded vesicles.

The half-lives and the respective kobs values were determined according to equations 3

and 4 in materials and methods. Results are shown in Table 7.

Table 7. kobs and half-life of C12 incorporated in PC liposomes in the presence of human plasma: influence of mean vesicle size

Formulation kobs x 103 (min

-1)

Half-life time

(min)

Ø (µm)

(PdI)

Non-Extruded 18.7 ± 1.5 37 ± 3 3.3

(0.7 - 0.8)

Extruded 29.5 ± 0.1 24 ± 1 0.2

(<0.2)

A higher half-life was observed for non-extruded (37 ± 3 min) in comparison to extruded

liposomes (24 ± 1 min). This may be explained due to the high number of lipid bilayers for non-

extruded vesicles. In contact with plasma, liposomes undergo adsorption of opsonins leading to

destabilization of lipid bilayers and release of incorporated material (Crommelin and Schreier,

1994; Moghimi and Hunter, 2001). In the present work, the release of C12 was followed by its

hydrolysis and consequent formation of POA. The higher number of lipid bilayers in non-

extruded liposomes will protect C12 from a faster hydrolysis rate.

B) C12 incorporated in DMPC liposomes

C12 was incorporated in DMPC, a neutral phospholipid with a moderate Tc of +23ºC and

thus presenting a higher rigidity than PC liposomes.

30

Two different liposome suspensions were prepared: non-extruded and extruded, with a

mean size of 6.5 and 0.2 µm, respectively. The stability of these two C12 formulations was

performed after incubation in human plasma and the hydrolysis of the prodrug and the formation

of POA were determined. Results are shown in Figure 16.

(a) (b)

Figure 16. Hydrolysis of C12 incorporated in DMPC liposomes and formation of POA in the presence of human plasma 50% (v/v). (a) Non-extruded liposomes. Initial concentration: 24 µM. (b) Extruded liposomes. Initial concentration: 60 µM. The results represent mean ± S.D. of three independent experiments

Again, as observed for PC liposomes, in Figure 15, a faster disappearance of C12 and

the respective formation of POA was observed for extruded liposomes (Figure 16 (b)) in

comparison with non-extruded vesicles (Figure 16 (a)). When C12 was incorporated in extruded

DMPC liposomes, 60 min after incubation with human plasma, a 75% reduction on initial

concentration of the prodrug was achieved. On the other hand for DMPC non-extruded vesicles,

after the same incubation period, only a 50% reduction on initial C12 concentration was

observed. Moreover, the hydrolysis rate in the following 2 h was slower and thus 180 min after

incubation with plasma more than 25% of the initial C12 concentration was still present.

The use of a lower initial C12 concentration for non-extruded liposomes reduced the

aggregation problems, during incubation period, associated to such large sized vesicles.

The half-lives and the respective kobs values were determined according to equations 3

and 4 in materials and methods. Results are shown in Table 8.

Table 8. kobs and half-life of C12 incorporated in DMPC liposomes in the presence of human plasma: influence of mean vesicle size

Formulation kobs x 103 (min

-1)

Half-Life Time

(min)

Ø (µm)

(PdI)

Non-Extruded 8.0 ± 1.0 88 ± 11 6.5

(0.3 – 0.4)

Extruded 29.4 ± 1.3 23 ± 2 0.20

(<0.2)

The use of a more rigid phospholipid, DMPC vs PC led to a decrease on hydrolysis rate

of C12 for non-extruded liposomes. In fact, the respective half-lives were 88 ± 11 and 37 ± 3

31

min. The mean sizes of these two non-extruded formulations were also very different: 6.5 and

3.3 µm for DMPC and PC liposomes, respectively. The obtained results confirm the hydrolysis

rate dependence on liposome mean sizes. For extruded vesicles no statistically significant

differences were observed between PC and DMPC formulations (p>0.05).

C) C12 incorporated in DMPC:DMPG liposomes

In order to study the influence of superficial charge of liposomes on C12 hydrolysis rate,

this prodrug was also incorporated in a lipid mixture containing, besides DMPC, a phospholipid

negatively charged, DMPG, with a Tc of +23ºC. Moreover, for this lipid composition,

DMPC:DMPG, the influence of mean size was studied using three different liposome

suspensions with diameters of 1.8 µm; 0.16 µm and 0.55 µm. The stability of these three C12

formulations was performed after incubation in human plasma and the hydrolysis of the prodrug

and the formation of POA were determined. Results are shown in Figure 17.

(a) (b)

(c)

Figure 17. Hydrolysis of C12 incorporated in DMPC:DMPG (7:3) liposomes and formation of POA in the presence of human plasma 50% (v/v). Initial concentration: 60 µM. (a) Non-Extruded liposomes with a mean size of 1.8 µm, (b) Extruded liposomes with a mean size of 0.16 µm, (c) Extruded liposomes with a mean size of 0.55 µm. The results represent mean ± S.D. of three independent experiments

32

A faster hydrolysis for C12 was observed for extruded liposomes as compared to non-

extruded vesicles. Moreover, the hydrolysis of C12 incorporated in both sized liposomes

showed similar profiles (Figures 17 (b) and (c)). Sixty min post-incubation these two

formulations have already hydrolyzed more than 75% of initial C12 while for non-extruded

vesicles the percentage of non-hydrolyzed C12 was higher than 36%.

The half-lives and the respective kobs values were determined according to equations 3

and 4 in materials and methods. Results are shown in Table 9.

Table 9. kobs and half-life of C12 incorporated in DMPC:DMPG (7:3) liposomes in the presence of human plasma: influence of mean vesicle size

Formulation kobs x 103 (min

-1)

Half-life time

(min)

Ø (µm)

(PdI)

Non-Extruded 19.4 ± 2.0 36 ± 4 1.7

(0.9 – 1)

Extruded 29.0 ± 1.0 24 ± 1 0.16

(<0.1)

Extruded 26.4 ± 0.2 26 ± 1 0.55

(0.4 – 0.5)

The overall results obtained for DMPC and DMPC:DMPG extruded formulations (Tables

8 and 9) did not reveal statistically significant differences in half-lives (p>0.05).

For non-extruded vesicles prepared with DMPC:DMPG a lower half-life was observed

when comparing to DMPC formulation. The presence of a negatively charged phospholipd in

the lipid composition enabled the achievement of smaller sized vesicles: 1.7 vs 6.5 µm. These

results are in accordance with literature, where the presence of negatively charged

phospholipids, such as DMPG, in the lipid composition reduces the aggregation of the vesicles

(Crommelin and Schreier, 1994). Data in Tables 8 and 9 clearly demonstrate a relationship

between mean size and hydrolysis rate.

D) C12 incorporated in DPPC

C12 was incorporated in DPPC, a neutral phospholipid with a Tc of +41ºC and

consequently presenting a higher rigidity than PC and DMPC previously tested. Two liposome

suspensions were prepared: non-extruded and extruded, with mean sizes of 3.8 µm and 1.8

µm, respectively. The stability of these two C12 formulations was performed after incubation in

human plasma and the hydrolysis of the prodrug and the formation of POA were determined.

Results are shown in Figure 18.

33

(a) (b)

Figure 18. Hydrolysis of C12 incorporated in DPPC liposomes and formation of POA in the presence of

human plasma 50% (v/v). (a) Non-extruded liposomes; Initial concentration: 24 µM (b) Extruded

liposomes. Initial concentration: 60 µM. The results represent mean ± S.D. of three independent

experiments

Again a faster decrease of C12 and the corresponding formation of POA was observed

for extruded liposomes (Figure 18 (b)) in comparison with non-extruded vesicles (Figure 18 (a)).

Sixty min post-incubation, extruded formulations already presented a 75% reduction of the initial

C12 concentration while for non-extruded vesicles the percentage of non-hydrolyzed C12 was

higher than 52%. Even 2 h post-incubation there were still more than 25% of the initial C12

concentration for non-extruded liposomes.

For extruded formulations, at the end of the analyzed stability period, the concentration

obtained for POA was higher than initial C12 values. The HPLC determinations of POA and C12

were performed separately and so these differences may be attributed to problems during

pippeting of samples.

The half-lives and the respective kobs values were determined according to equations 3

and 4 in materials and methods. Results are shown in Table 10.

Table 10. kobs and half-life of C12 incorporated in DPPC liposomes in the presence of human plasma: influence of mean vesicle size

Formulation kobs x 103 (min

-1)

Half-life time

(min)

Ø (µm)

(PdI)

Non-Extruded 9.7 ± 1.7 75 ± 9 3,8

(0.9 - 1)

Extruded 26.5 ± 0.5 26 ± 1 1.8

(0.4 – 0.5)

For extruded vesicles no statistically significant differences were observed in terms of

half-life for PC, DMPC and DPPC formulations (p>0.05). Besides the superior mean size of

DPPC extruded formulation, 1.8 µm, they presented a very high PdI (around 0.5).

Non-extruded DPPC liposomes presented a C12 half-life of 75 ± 9 min. This value is

similar to the one obtained for DMPC 88 ± 11 min (p>0.05) and higher for PC 37 ± 3 min.

34

Statistically significant differences were observed between PC vs DMPC and DPPC liposomes

(p<0.05).

Taking into account the overall results, two main factors are responsible for hydrolysis

rate of C12: the mean size and the rigidity of liposomes.

E) C12 incorporated in DPPC:DPPG

The influence of superficial charge of liposomes on C12 hydrolysis rate was also

evaluated by incorporating this prodrug in a lipid mixture containing, besides DPPC, a

negatively charged phospholipid, the DPPG, with a Tc of +41ºC. In this study the stability of C12

formulations was only evaluated for non-extruded vesicles. Liposomes presented a mean size

of 0.96 µm. The stability of this formulation was performed after incubation in human plasma

and the hydrolysis of the prodrug and the formation of POA were determined. Results are

shown in Figure 19.

Figure 19. Hydrolysis of C12 incorporated in DPPC:DPPG (7:3) non-extruded liposomes and formation of POA in the presence of human plasma 50% (v/v). Initial concentration: 24 µM. The results represent mean ± S.D. of three independent experiments

According to the obtained results, the reduction of C12 was followed by a concomitant

increase on POA. One hundred and eighty min post-incubation less than 3 % of C12 was in

non-hydrolyzed form while around 97% was converted in POA.

The half-life and the respective kobs value were determined according to equations 3

and 4 in materials and methods. Results are shown in Table 11.

Table 11. kobs and half-life of C12 incorporated in DPPC:DPPG (7:3) non-extruded liposomes in the presence of human plasma

Formulation kobs x 103 (min

-1)

Half-life time (min)

Ø (µm) (PdI)

Non-Extruded 22.2 ± 2.1 32 ± 3 0.96

(0.4 – 0.5)

35

The inclusion of DPPG in the lipid composition resulted in a faster C12 hydrolysis when

compared to DPPC: 32 ± 3 min and 75 ± 9 min (Tables 10 and 11). The same effect has been

already observed for DMPC vs DMPC:DMPG with a reduction from 88 ± 11 min to 36 ± 4 min.

The presence of a negatively charged phospholipid in the lipid bilayer has lead to a reduction on

the mean size of liposomes and concomitant decrease on the C12 half-life in presence of

human plasma.

Although extruded vesicles were not performed for the lipid composition DPPC:DPPG

we may expect that an hydrolysis rate for C12 should be similar to the one observed for

DMPC:DMPG extruded liposomes (24 ± 1 min).

F) C12 incorporated in DMPC:DSPE-PEG

The presence of PEG in the lipid composition on C12 hydrolysis rate was also

evaluated. As widely described in literature, PEG is able to reduce plasmatic protein adsorption

at liposome surface when included in lipid mixtures (Allen and Cullis, 2004; Dos Santos et al.,

2007; Torchilin, 2005). The stability of this formulation was performed after incubation in human

plasma and the hydrolysis of the prodrug and the formation of POA were determined. Results

are shown in Figure 20.

Figure 20. Hydrolysis of C12 incorporated in DMPC:DSPE-PEG (2.85:0.15) extruded liposomes and formation of POA in the presence of human plasma 50% (v/v). Initial concentration: 60 µM. Extruded liposomes. The results represent mean ± S.D. of three independent experiments

According to the obtained results the reduction of C12 was followed by a concomitant

increase on POA. Sixty min post-incubation a reduction of more than 75% of the initial C12

concentration was achieved. The percentage of non-hydrolyzed C12 was 21% while around

79% was converted in POA.

The half-life and kobs were determined according to equations 3 and 4 in materials and

methods. Results are shown in Table 12.

36

Table 12. kobs and half-life of C12 incorporated in DMPC:DSPE-PEG (2.85:0.15) liposomes in the presence of human plasma

Formulation kobs x 103 (min

-1)

Half-life time (min)

Ø (µm) PdI

Extruded 26.8 ± 0.4 26 ± 1 0.15

(<0.2)

The half-life of C12 obtained for liposomes containing DSPE-PEG, in the lipid

composition, presented similar results to all the formulations submitted to extrusion procedures

(see tables 7-10).

In order to summarize all obtained data related with C12 stability in the presence of

plasma in Figure 21 are shown half-lives for all C12 studied liposomal formulations.

A B C D E F G H I J

100

80

60

40

20

0

Non-extruded liposomes Extruded liposomes

Ha

lf-l

ife

(m

in)

Figure 21. Half-life of C12 formulations after incubation in human plasma 50% (v/v): A – non-extruded PC

liposomes; B – non-extruded DMPC liposomes; C – non-extruded DPPC liposomes; D – non-extruded

DMPC:DMPG (7:3) liposomes; E – non-extruded DPPC:DPPG liposomes; F –extruded PC liposomes; G –

extruded DMPC liposomes; H – extruded DPPC liposomes;I – DMPC:DMPG (7:3) extruded liposomes; J –

DMPC:DSPE-PEG (2.85:0.15) extruded liposomes

In order to evaluate the parameters able to influence the hydrolysis rate of C12 in the

presence of human plasma, a systematic study was performed. In this context several

conditions were studied: composition and rigidity of the lipid bilayer, mean size and superficial

charge of the vesicles.

Taking into account the overall results it is clear that the main factors able to influence

the hydrolysis rate of C12 liposomal formulations in the presence of human plasma are the

mean size, the rigidity and the presence of negatively charged phospholipids in the lipid mixture.

The higher half-life values were observed for non-extruded formulations prepared with

neutral phospholipids that also represented the larger liposomes: DMPC and DPPC with 88 ±

11 min and 75 ± 9 min, respectively. In a previous work, similar results were observed (Simões,

2005). However, non-extruded liposomes are very heterogeneous and thus from a

37

pharmaceutical point of view are not acceptable for parenteral administration. In addition, their

heterogeneity does not allow the achievement of a reproducible formulation from batch to batch.

Intermediate half-life values ranging from 32 to 37 min were obtained for non-extruded

liposomes prepared with fluid (PC) or negatively charged phospholipid (DMPC:DMPG and

DPPC:DPPG).

For extruded liposomes the half-life ranged from 23 to 26 min independent of rigidity

and of the presence of negatively charged phospholipids.

In the presence of biological fluids liposomes suffer the adsorption of plasmatic proteins

at their surface leading to destabilization of their lipid bilayers. The high number of lamella as it

occurs in non-extruded liposomes is the main reason why these type of liposomes presented

lower hydrolysis rates for the incorporated prodrug. These observations are in accordance with

previous reports (Hernandezcaselles et al., 1993; Ishida et al., 2002). When C12 in free form

was incubated in human plasma, the half-life value determined ranged from 38 to 43 min (Table

6) evidencing a lower hydrolysis rate as compared to C12 extruded liposomes. Nevertheless,

the parenteral administration of a prodrug with hydrophobic properties, such as C12 in free

form, requires the use of solubilizing agents that may lead to toxic side effects. Liposomes,

particularly extruded vesicles, are able to solve solubility problems and thus they are preferable

for being tested in in vivo models, as a high reproducibility between batches is achieved

(Crommelin and Schreier, 1994). Moreover, sometimes in vitro studies are not able to correlate

exactly what happens in in vivo (Yardley and Croft, 2000).

3.4.3.3. Stability of C12 formulations – Influence of C12 concentration

C12 Hydrolysis rate

All stability studies above presented were performed using C12 concentrations ranging

from 24 to 60 µM. In order to simulate the in vivo conditions, the C12 hydrolysis rate was also

evaluated using the same therapeutic dose that was administered in the M. avium murine model

of infection: 1300 µM in human plasma 50% (v/v). A C12 liposomal formulation prepared with

the lipid mixture DMPC:DMPG with a mean size below 0.2 µm was used. The hydrolysis rate of

C12 in the free form and incorporated in liposomes was compared. Results are shown in Figure

22.

38

(a) (b)

Figure 22. Hydrolysis of C12 and formation of POA after incubation with human plasma 50% (v/v). Tested formulations: C12 in the free form (a) and incorporated in DMPC:DMPG (7:3) extruded liposomes (b) Initial concentration: 1300 µM. The results represent mean ± S.D. of three independent experiments

A lower hydrolysis rate of C12 and the respective formation of POA was observed for

both formulations as compared to the above tested formulations (Figures 13 (b) and 17 (b))

using lower initial C12 concentrations. The decrease of C12 was observed for more than 400

min.

Although the initial C12 concentration was the same for both formulations, for C12

liposomes this value was around 1200 µM, while for free C12 was below 1000 µM. This

decrease may be explained due to a possible precipitation of free C12 in the presence of

plasma proteins. The use of a high concentration of a lipophilic molecule such as C12, even

using 20% of ACN in the reaction medium, may not be enough to avoid precipitation. In fact, the

quantification of POA for free C12 reached higher values than the correspondent initial C12

concentration: approximately1200 µM 400 min post-incubation in human plasma. In addition,

the same effect was observed for C12 in the liposomal form.

Transposing these results for an in vivo model and using this lipid composition, 400 min

after parenteral administration a high fraction of this formulation will no longer be in

bloodstream. The removal from blood circulation by the cells of MPS and accumulation,

particularly in liver and spleen, is widely described in literature for this type of liposomes (Allen

and Hansen, 1991; Allen and Cullis, 2004; Torchilin, 2005;). This natural accumulation has been

exploited for efficiently delivering of antibiotics to these tissues for treatment of intracellular

infections (Fielding and Lasic, 1999; Gaspar et al., 2008a; Gaspar et al., 2008b; Pinto-

Alphandary et al., 2000).

Half-lives and kobs were determined according to equations 3 and 4 in materials and

methods (Table 13).

39

Table 13. kobs and half-life of C12 in free form and incorporated in DMPC:DMPG (7:3) extruded liposomes in the presence of human plasma

C12 kobs x 103 (min

-1)

Half-life time

(min)

Free form 4.9 ± 0.5 95 ± 5

Liposomal formulation 7.3 ± 0.4 144 ± 13

As already mentioned, using an initial concentration of C12 ranging from 24 to 60 µM

only for non-extruded liposomes prepared with DMPC and DPPC a higher half-life for C12 was

observed as compared to free prodrug (see Figure 21 and Table 6): 75 ± 9 and 88 ± 11 min vs

43 ± 4 min, respectively. On the other hand, when C12 was incorporated in extruded liposomes

prepared with DMPC:DMPG the respective half-life obtained was 24 ± 1 min (Table 9). An

increase on the initial C12 concentration in the reaction medium led to the achievement of a

higher half-life for C12 in DMPC:DMPG extruded liposomes than the one obtained for C12 in

the free form: 144 ± 13 and 95 ± 5 min, respectively. In this latter experimental condition plasma

proteins were not enough to destabilize the total amount of liposomes present in the reaction

medium (Hernandezcaselles et al., 1993; Ishida et al., 2002).

3.4.3.4. Stability of liposome structure in presence of human plasma

The evaluation of physicochemical properties of liposomes in the presence of human

plasma using a 1300 µM C12 concentration, the same as later used in the M. avium murine

model of infection, was performed. These studies intended to extrapolate if liposomes maintain

their physicochemical properties after i.v. administration to mice. For this study, C12 was

incorporated in DMPC:DMPG extruded liposomes following their incubation in human plasma at

37ºC. At defined times aliquots of suspensions were taken and applied onto the top of a

Sephadex G200 column. Eluted fractions (1 mL) were recovered and the absorbance of all

samples was recorded by spectrophotometry at 420 nm. In Figure 23 is shown the graphical

representation of the fractions eluted from columns at time zero and 1 and 3 h after liposomes

incubation.

Figure 23.Absorbance at 420 nm of eluted C12 DMPC:DMPG extruded liposomes following their application on the top of a Sephadex G200 column. The results represent mean ± S.D. of three independent experiments

40

As shown in Figure 23 a slight reduction on the fraction of liposomes was observed for

samples recovered 1 and 3 h post-incubation. This slight decrease on the absorbance is in

accordance with the C12 hydrolysis using the higher concentration (1300 µM) (Figure 22 (b)).

Moreover, the determination of mean size for the eluted fractions corresponding to liposomes

demonstrated a slight decrease from 0.16 to 0.12 µm (Table 14). These results evidence that

the incubation of liposomes in human plasma led to a reduction on the number of lipid bilayers

and a concomitant release of incorporated C12 followed by its hydrolysis. Nevertheless, this

study confirms the high stability of this lipid composition when in presence of human plasma.

Table 14. Stability of liposome structure in the presence of human plasma - Mean size of liposomes at time zero and 1 and 3h post-incubation following their application on the top of a Sephadex G200 column

Time post incubation (h) Ø (µm) / (PdI)

0 0.16 / (<0.2)

1 0.12 / (<0.2)

3 0,12 / (<0.2)

Lipid composition – extruded DMPC:DMPG

3.5. Mycobacterium avium murine model of infection

3.5.1. Biological evaluation of antimycobacterial formulations

Previous in vitro studies using C12 evidenced antimycobacterial activity against M.

tuberculosis (Simões, 2005; Simões et al., 2009). With the aim to evaluate the possible

antimycobacterial effect of C12 against M. avium some preliminary studies were performed. To

our knowledge it was the first time that this in vivo experiment was assessed. According to

literature, M. avium is intrinsically resistant to PZA (Zhang and Mitchison, 2003) and thus it is of

great interest to elucidate if new synthesized esters of POA are able to overlap this handicap.

The M. avium strain (DSMZ 44157) was chosen as this microorganism is a level 2

pathogenic biological agent, whereas M. tuberculosis must be handled in facilities with biosafety

level P3.

Before the establishment of the animal model the exact quantification of the inoculum of

M. avium DSMZ 44157 and the in vivo evolution of mycobacterial infection were carried out.

3.5.2. In vivo evolution of mycobacterial infection

Mycobacterial murine models of infection can affect different organs such as lungs,

spleens, livers and lymph nodes. This is dependent not only on the properties of the inocula

used but also on the route selected for the establishment of the infection (Cynamon and

DeStefano, 1999).

41

In order to evaluate which are the main infected organs, a preliminary study was

performed. For this purpose, BALB/c mice were injected intravenously with an inoculum of M.

avium DSMZ 44157 corresponding to 5x105 colony forming units (CFU). Fifteen days after

infection mice were sacrificed and liver, spleen and lungs were removed and the number of

CFU were determined (Gaspar et al, 2000). In Figure 24 are represented the CFUs per g of

organ in the analyzed time point.

15 days after infection induction

108

106

104

102

CF

U /

g o

rga

nLiver Spleen Lung

Figure 24. Evolution of CFU per g of organ in liver, spleen and lung of BALB/c mice 15 days after infection induction. Mice received an i.v. injection of an inoculum of M. avium DSMZ 44157. Infection dose: 5x10

5

CFU per mouse. Results are expressed as mean ± S.D. of 4 mice

The obtained results indicate that the main infected organs are liver and spleen. The

lung was the less affected organ with a number of CFU per g of organ below 1x105.

Taking these results into account the biological evaluation exerted by the selected

formulations was determined in liver and spleen.

3.5.3. Influence of antimycobacterial formulations on M. avium murine

model

The murine model of infection intended to investigate if in vitro stability studies of C12

formulations, performed in the presence of plasma, could be correlated with their therapeutic

response. With this purpose, the lipid composition DMPC:DMPG was chosen to be tested in the

M. avium murine model of infection. The prodrug C12 was incorporated in extruded and non-

extruded liposomes. In Table 15 are shown some physicochemical properties of the liposomal

formulations used in treatment schedule.

Table 15. C12 liposomal formulations used for the treatment of M. avium murine model of infection

Formulation

Lipid Composition

(Molar Ratio)

Extruded / Non-

Extruded

Ø (µm)

(PdI)

Zeta Potential

(mV)

17 DMPC:DMPG (7:3) Extruded 0.15

(<0.2) -29 ± 3

18 DMPC:DMPG (7:3) Non-Extruded* 1.2

(0.7 – 0.8) -32 ± 2

Ø – mean size of liposomes; PdI – Polydispersity index. *This formulation was extruded through filter of 2 µm of porosity

42

Two control groups were used: infected and non treated animals (Cont.I and Cont.II)

Cont.I corresponds to the bacterial loads in liver and spleen of mice in the beginning of the

treatment, while Cont.II represents the bacterial loads at the end of treatment schedule. Two

other groups received PZA or POA in the free form. In Figure 25 are represented the number of

CFU per g of organ for these tested groups.

**statistically significant from Cont.II

Figure 25. Evolution of M. avium infection in mice. Mice received an i.v. injection of an inoculum of M. avium strain DSMZ 44157. Infection dose: 1x10

6 CFU per mouse. Treatment started two weeks after

infection. Mice received i.v. injections of PZA and POA three times a week for two weeks. Administered dose: 12 mg/kg of body weight. Control I (Cont.I) corresponds to the bacterial load in the beginning of treatment; the other groups correspond to the bacterial loads at the end of treatment: Control II (Cont.II) – untreated animals; POA – Pyrazinoic Acid; PZA – Pyrazinamide. Results are expressed as mean ± S.D. of at least 5 mice per group.

The results indicate a high progression on the mycobacterial infection. Cont.I presented

a number of CFU/g of organ of around 108, while two weeks after infection a 10 fold increase on

the bacterial load was observed (Cont.II). For mice receiving PZA and POA no statistically

significant differences in bacterial loads in liver were detected in comparison with Cont.II

(p>0.4). One the other hand, in spleen a therapeutic effect for both molecules was achieved.

Statistically significant differences were observed (p<0.05) in both cases.

C12 was administered in free and liposomal forms. In Figure 26 are shown the number

of CFU per g of organ of all analyzed animal groups two weeks after treatment: untreated

animals (Cont.II) and treated animals (POA, C12, LIP1 and LIP2).

43

* Statistically significant from Cont.II; ** Statistically significant from Cont.II

Figure 26. Evolution of M. avium infection in mice. Influence of administered formulations on CFU per g of organ. Mice received an i.v. injection of an inoculum of M. avium strain DSMZ 44157. Infection dose: 1x10

6

CFU per mouse. Treatment started two weeks after infection. Mice received i.v. injections of formulations under study three times a week for two weeks. Administered dose: 12 mg/kg of body weight. Studied formulations: Control II (Cont.II) – untreated animals; POA – Pyrazinoic Acid; C12 – Prodrug in the free form; LIP1 – C12 incorporated in DMPC:DMPG (7:3) non-extruded liposomes; LIP2 – C12 incorporated in DMPC:DMPG (7:3) extruded liposomes. Results are expressed as mean ± S.D. of at least 4 mice per group

The results show, as already mentioned, that for mice treated with POA the degree of

infection in liver was very similar to Cont.II (p>0.05) while in spleen a significant reduction was

observed (p<0.05). The administration of C12 in the free and liposomal forms (extruded

liposomes – LIP2) reduced significantly the bacterial loads either in liver or in spleen (around 10

fold) when compared to Cont.II (p<0.05). On the opposite, the use of non-extruded liposomes

(LIP1) did not show a therapeutic response, as no statistical differences over Cont.II were

observed (p>0.4 and p>0.1 for spleen and liver, respectively). These results demonstrate that in

vitro studies are not always able to correlate exactly what happens in in vivo (Yardley and Croft,

2000). Indeed, the increase of around 30% in the half-life observed for LIP1 vs LIP2 (Figure 21)

in plasma stability assays was not accomplished by an increase on the therapeutic effect.

When comparing bacterial loads in liver for mice treated with POA vs LIP2, the

differences in the number of CFU/g organ were statistically significant (p<0.05), but not in

spleen (p>0.08). Moreover, for mice groups treated with POA, C12 and LIP2, no statistically

significant differences were observed in infection degree in spleen (p>0.4). Based on these

results, the optimization of a liposomal formulation able to exert a superior therapeutic effect in

this organ should be assayed.

In this in vivo model the degree of infection of all animals was very high. Thus, the

possibility of achieving higher reductions on bacterial loads is more difficult. In addition the M.

avium strain used in the present thesis is described in literature as being more resistant to

several antibiotics than some M. tuberculosis strains (Inderlied et al., 1993; Luo et al., 2011).

The performance of other M. avium murine models of infection using more susceptible strains

should be considered in order to evaluate if the therapeutic effect is even higher than the one

already obtained.

44

These in vivo studies exhibit very promising results. As already mentioned, M. avium is

intrinsically resistant to PZA and this may be due to an efficient POA efflux mechanism (Wade

and Zhang, 2004; Zhang and Mitchison, 2003). However, a therapeutic effect of POA and PZA

was achieved for the spleen. For C12 formulations no statistically significant differences were

observed, in terms of bacterial loads either in liver or in spleen. The use of lower administered

doses should be considered in further in vivo tests in order to clarify these therapeutic

responses.

The similar antimycobacterial effect of C12 formulations (C12 vs LIP2) could be a result

of a plateau in terms of dose response. According to literature, in other animal models this effect

has also been observed when high therapeutic doses are administered (Lala et al., 2006;

Sarkar et al., 2002). Taking this into account, the use of lower doses could lead to statistically

significant differences between free and liposomal C12, showing the therapeutic advantages of

using liposomes, based on the assumption of different biodistribution profiles.

45

4. Conclusions and Future Perspectives

Mycobacterial species resist to a variety of antimycobacterial agents because of their

thick cell wall that confers an exceptionally strong barrier to antibiotics. In an attempt to overlap

these drawbacks a viable strategy may be achieved by the synthesis of new molecules or

chemical modification of the already available ones. In particular, the prodrug approach via

activation inside mycobacterial cells is a relevant alternative especially considering the

problematic penetration of drugs into these cells (Valente et al., 2011).

PZA, a first line agent for treatment of TB, is also a prodrug. In order to exert

antimycobacterial activity, PZA requires activation by the Mycobacterium enzyme

pyrazinamidase (PZase) to be transformed into its active form, the POA. This enzyme

conveniently activates the drug only inside the bacteria (Pires, 2011). However, since the

activation of PZA is only dependent on one enzyme, it has been observed the emergence of

resistant strains to this prodrug. To overcome these drawbacks the synthesis of new POA

prodrugs were conducted by several authors where some compounds displayed higher activity

than PZA against M. tuberculosis, M. avium and M. kansasii (Cynamon et al., 1992; Cynamon

et al., 1995). However, these new molecules displayed low stability in presence of biological

fluids (Bergmann et al., 1996). Simões and co-workers have synthesized series of POA

prodrugs with different alkoxy chain groups (Simões, 2005; Simões et al., 2009; Valente et al.,

2011). They found that longer fatty acid chains were more resistant to plasma hydrolysis. As

these prodrugs are easily activated by mycobacterial esterases, this may contribute to a

reduction on the occurrence of resistances against mycobacterial strains. In in vitro tests some

POA esters were found to be 10 times more active against sensitive M. tuberculosis than PZA

(Simões et al., 2009).

Liposomal formulations of two POA esters, C4 and C12, were developed and their

therapeutic potential, in a preliminary in vivo model infected with Mycobacterium avium DSMZ

44157, was evaluated. The rational for associating these POA esters to liposomes was based

on the ability of these lipid systems to circumvent some solubility problems associated to these

molecules and, after parenteral administration, they can passively target liver and spleen, the

main affected organs in M. avium infections.

The physicochemical characterization of liposomal formulations enabled the

achievement of incorporation efficiencies higher than 90% for C12 while very low values were

obtained for C4 (below 6%). C12 was then selected for further in vitro and in vivo studies. The

log Poct of C12 is around 4.55, owning a hydrophobic character. Due to this property, it is

hypothesized that this prodrug may be accommodated in the lipid bilayer (Allen and Stuart,

1999; Defrise-Quertain et al., 1984;). This assumption was confirmed by calorimetric studies as

an increase on the enthalpy was observed for loaded liposomes in comparison with the

unloaded ones.

Stability studies of C12 liposomes in HEPES buffer evidenced the high stability of these

formulations considering the percentage of incorporated prodrug and the preservation of non-

46

hydrolyzed form. A systematic study of C12 formulations in human plasma was also performed.

C12 in free form presented a half-life of around 43 ± 4 min. Almost all liposomal formulations

presented higher hydrolysis rates than C12 in the free form, with the exception for neutral and

non-extruded liposomes. However, from the pharmaceutical point of view, it is crucial the

achievement of liposomes with a high homogeneous mean size and the preservation of all

physicochemical properties as they will influence the in vivo profile of the correspondent

formulation. Taking this into account, extruded liposomes appear more attractive as the

maintenance of their characteristics and the reproducibility from batch to batch was observed.

The influence of C12 initial concentration on the hydrolysis rate was also evaluated, evidencing

that, when similar experimental conditions as those tested in the in vivo model were used,

higher stability for C12 in extruded liposomes was achieved when compared to the respective

free form. These observations are in accordance with literature as plasma proteins are not able

to destabilize the total amount of liposomes in reaction medium (Hernandezcaselles et al.,

1993; Ishida et al., 2002).

With the aim to evaluate the possible antimycobacterial effect of C12 against a M.

avium strain DSMZ 44157, some preliminary studies were performed. From our knowledge, it

was the first time that this in vivo experiment was assayed. Taking into consideration the

promising in vitro results obtained for POA ester prodrugs against M. tuberculosis (Simões et

al., 2009) we intended to evaluate whether the selected C12 formulations could also overcome

the intrinsic resistance of M. avium to PZA (Zhang and Mitchison, 2003). Even using a M. avium

strain resistant to most antibiotics (Inderlied et al., 1993; Luo et al., 2011), it was possible to

observe a therapeutic effect for mice treated with C12, both in free form and incorporated in

extruded liposomes. Inexplicably, mice groups receiving POA and PZA also demonstrated a

reduction on the bacterial loads in spleen. The effect may be due to the high administered dose.

These preliminary in vivo tests constitute good future prospects and so the use of other

treatment schedules and mycobacterial strains may be considered for future work. Moreover,

the performance of biodistribution studies should also be assayed to evaluate the influence of

doses and lipid compositions, to select the ones that would lead to a higher prodrug

accumulation within the infected organs. Nevertheless, these preliminary in vivo results may be

envisioned as an alternative strategy for treatment of mycobacterial infections.

47

5. References

Allen T.M. and Cullis P.R. (2004) Drug Delivery Systems: Entering the Mainstream. Science 303: pp 1818-1822. Allen T.M. and Hansen C. (1991) Pharmacokinetics of Stealth Versus Conventional Liposomes - Effect of Dose. Biochimica et Biophysica Acta 1068: pp 133-141. Allen T.M., Newman M.S., Woodle M.C., Mayhew E. and Uster P.S. (1995) Pharmacokinetics and Antitumor-Activity of Vincristine Encapsulated in Sterically Stabilized Liposomes. International Journal of Cancer 62: pp 199-204. Allen T.M. and Stuart D.D. (1999) Liposome Pharmacokinetics: classical, sterically stabilized, cationic liposomes and immunoliposomes, in Liposomes: Rational Design (Janoff A.S. ed) pp 63-87, Marcel Dekker, Inc, New York, Basel. Bakker-Woudenberg I.A.J.M., Lokerse A.F., Tenkate M.T., Mouton J.W., Woodle M.C. and Storm G. (1993) Liposomes With Prolonged Blood-Circulation and Selective Localization in Klebsiella-Pneumoniae Infected Lung-Tissue. Journal of Infectious Diseases 168: pp 164-171. Bangham A.D., Standish M.M. and Watkins J.C. (1965) Diffusion of Univalent Ions Across Lamellae of Swollen Phospholipids. Journal of Molecular Biology 13: pp 238-&. Bergmann K.E., Cynamon M.H. and Welch J.T. (1996) Quantitative Structure-Activity Relationships for the in Vitro Antimycobacterial Activity of Pyrazinoic Acid Esters. Journal of Medicinal Chemistry 39: pp 3394-3400. Bolotin E.M., Cohen R., Bar L.K., Emanuel S.N., Ninio S., Lasic D.D. and Barenholz Y. (2012) Ammounium Sulfate Gradients for Efficient and Stable Remote Loading of Amphipathic Weak Bases into Liposomes and Ligandoliposomes. Journal of Liposome Research 4: pp 455-479. Carvalheiro M., Jorge J., Eleuterio C., Pinhal A.F., Sousa A.C., Morais J.G. and Cruz M.E.M. (2009) Trifluralin Liposomal Formulations Active Against Leishmania Donovani Infections. European Journal of Pharmaceutics and Biopharmaceutics 71: pp 292-296. Castelli F., Caruso S. and Uccella N. (2003) Effect of Simple Olive Biophenols and Analogues on the Thermotropic Behavior of Biological Model Membranes. European Journal of Lipid Science and Technology 105: pp 260-265. Constantino L., Cruz M.E.M., Mehta R. and Lopez-Berestein G. (1993) Formulation and Toxicity of Liposomes Containing Rifampicin. Journal of Liposome Research 2: pp 275-301. Cook J.L. (2010) Nontuberculous Mycobacteria: Opportunistic Environmental Pathogens for Predisposed Hosts. British Medical Bulletin 96: pp 45-59. Corvo M.L., Jorge J.C.S., van't Hof R., Cruz M.E.M., Crommelin D.J.A. and Storm G. (2002) Superoxide Dismutase Entrapped in Long-Circulating Liposomes: Formulation Design and Therapeutic Activity in Rat Adjuvant Arthritis. Biochimica et Biophysica Acta-Biomembranes 1564: pp 227-236. Cosma C.L., Sherman D.R. and Ramakrishnan L. (2003) The Secret Lives of the Pathogenic Mycobacteria. Annual Review of Microbiology 57: pp 641-676. Crommelin D.J.A., Grit M., Talsma H. and Zuidam N.J. (1994) Liposomes As Carriers for Drugs and Antigens - Approaches to Preserve Their Long-Term Stability. Drug Development and Industrial Pharmacy 20: pp 547-556. Crommelin D.J.A. and Schreier H. (1994) Liposomes, in Colloidal Drug Delivery Systems (Kreuter J. ed) pp 73-190, Marcel Dekker, Inc, New York. Cruz M.E.M., Simões S.I., Corvo M.L., Martins M.B.F. and Gaspar M.M. (2009) Formulation of NPDDS for Macromolecules, in Drug Delivery Nanoparticles: Formulation and Characterization (Yashwant Pathak DT ed) pp 35-49, Informa Healthcare. Cynamon M.H. and DeStefano M.S. (1999) Beige Mouse Model of Disseminated Mycobacterium avium Complex Infection, in Handbook of Animal Models of Infection Academic Press.

48

Cynamon M.H., Gimi R., Gyenes F., Sharpe C.A., Bergmann K.E., Han H.J., Gregor L.B., Rapolu R., Luciano G. and Welch J.T. (1995) Pyrazinoic Acid-Esters With Broad-Spectrum In-Vitro Antimycobacterial Activity. Journal of Medicinal Chemistry 38: pp 3902-3907. Cynamon M.H., Klemens S.P., Chou T.S., Gimi R.H. and Welch J.T. (1992) Antimycobacterial Activity of A Series of Pyrazinoic Acid-Esters. Journal of Medicinal Chemistry 35: pp 1212-1215. Defrise-Quertain F., Chatelain P., Delmelle M. and Ruysschaert M.J. (1984) Model Studies for Drug Entrapment and Liposome Stability, in Liposome Technology - Incorporation of Drugs, Proteins and Genetic Materials (Gregoriadis G ed) pp 1-17, CRC Press, Inc, Boca Raton, Florida, USA. Demetzos C. (2008) Differential Scanning Calorimetry (DSC): A Tool to Study the Thermal Behavior of Lipid Bilayers and Liposomal Stability. Journal of Liposome Research 18: pp 159-173. Dos Santos N., Allen C., Doppen A.M., Anantha M., Cox K.A.K., Gallagher R.C., Karlsson G., Edwards K., Kenner G., Samuels L., Webb M.S. and Bally M.B. (2007) Influence of Poly(Ethylene Glycol) Grafting Density and Polymer Length on Liposomes: Relating Plasma Circulation Lifetimes to Protein Binding. Biochimica et Biophysica Acta-Biomembranes 1768: pp 1367-1377. du Toit L.C., Pillay V. and Danckwerts M.P. (2006) Tuberculosis Chemotherapy: Current Drug Delivery Approaches. Respiratory Research 7. Fernandes J.P.S., Pasqualoto K.F.M., Felli V.M.A., Ferreira E.I. and Brandt C.A. (2010) QSAR Modeling of a Set of Pyrazinoate Esters As Antituberculosis Prodrugs. Archiv der Pharmazie 343: pp 91-97. Fielding R.M. and Lasic D.D. (1999) Liposomes in the Treatment of Infectious Diseases. Expert Opinion on Therapeutic Patents 9: pp 1679-1688. French A.L., Benator D.A. and Gordin F.M. (1997) Nontuberculous Mycobacterial Infections. Medical Clinics of North America 81: pp 361-&. Gaspar M.M., Boerman O.C., Laverman P., Corvo M.L., Storm G. and Cruz M.E.M. (2007) Enzymosomes With Surface-Exposed Superoxide Dismutase: In Vivo Behaviour and Therapeutic Activity in a Model of Adjuvant Arthritis. Journal of Controlled Release 117: pp 186-195. Gaspar M.M., Cruz A., Fraga A.G., Castro A.G., Cruz M.E.M. and Pedrosa J. (2008a) Developments on Drug Delivery Systems for the Treatment of Mycobacterial Infections. Current Topics in Medicinal Chemistry 8: pp 579-591. Gaspar M.M., Cruz A., Penha A.E., Reymao J., Sousa A.C., Eleuterio C.V., Domingues S.A., Fraga A.G., Filho A.L., Cruz M.E.M. and Pedrosa J. (2008b) Rifabutin Encapsulated in Liposomes Exhibits Increased Therapeutic Activity in a Model of Disseminated Tuberculosis. International Journal of Antimicrobial Agents 31: pp 37-45. Gaspar M.M., Neves S., Portaels F., Pedrosa J., Silva M.T. and Cruz M.E.M. (2000) Therapeutic Efficacy of Liposomal Rifabutin in a Mycobacterium Avium Model of Infection. Antimicrobial Agents and Chemotherapy 44: pp 2424-2430. Gaspar M.M., PerezSoler R. and Cruz M.E.M. (1996) Biological Characterization of L-Asparaginase Liposomal Formulations. Cancer Chemotherapy and Pharmacology 38: pp 373-377. Georgiev S.V. (1994) Developmental Therapeutics of Mycobacterium Avium Complex (MAC) Infections. International Antimicrobial Agents 4: pp 157-173. Gregoriadis G. and Ryman B.E. (1972) Fate of Protein-Containing Liposomes Injected Into Rats - Approach to Treatment of Storage Diseases. European Journal of Biochemistry 24: pp 485-&. Gunstone F.D., Padley F.B. and Harwood J.L. (1986) In the Lipid Handbook. Chapman and Hall, London. Gupta D., Gupta S.V., Lee K.D. and Amidon G.L. (2009) Chemical and Enzymatic Stability of Amino Acid Prodrugs Containing Methoxy, Ethoxy and Propylene Glycol Linkers. Molecular Pharmaceutics 6: pp 1604-1611. Hernandezcaselles T., Villalain J. and Gomezfernandez J.C. (1993) Influence of Liposome Charge and Composition on Their Interaction With Human Blood-Serum Proteins. Molecular and Cellular Biochemistry 120: pp 119-126.

49

Inderlied C.B., Kemper C.A. and Bermudez L.E.M. (1993) The Mycobacterium-Avium Complex. Clinical Microbiology Reviews 6: pp 266-310. Ishida T., Harashima H. and Kiwada H. (2002) Liposome Clearance. Bioscience Reports 22: pp 197-224. Konno K., Feldmann F.M. and McDermot.W. (1967) Pyrazinamide Susceptibility and Amidase Activity of Tubercle Bacilli. American Review of Respiratory Disease 95: pp 461-&. Lala S., Gupta S., Sahu N.P., Mandal D., Mondal N.B., Moulik S.P. and Basu M.K. (2006) Critical Evaluation of the Therapeutic Potential of Bassic Acid Incorporated in Oil-in-Water Microemulsions and Poly-D,L-Lactide Nanoparticles Against Experimental Leishmaniasis. Jounal of Drug Targeting 14 (4): pp 171-179. Liu J.B., Lee H., Huesca M., Young A.P. and Allen C. (2006) Liposome Formulation of a Novel Hydrophobic Aryl-Imidazole Compound for Anti-Cancer Therapy. Cancer Chemotherapy and Pharmacology 58: pp 306-318. Luo X., Pires D., Ainsa J.A., Gracia B., Mulhovo S., Duarte A., Anes E. and Ferreira M.J.U. (2011) Antimycobacterial Evaluation and Preliminary Phytochemical Investigation of Selected Medicinal Plants Traditionally Used in Mozambique. Journal of Ethnopharmacology 137: pp 114-120. Maskill H. (1989) The Physical Basis of Organic Chemistry. Press, U.K. Miller J.N. (1991) Basic Statistical-Methods for Analytical-Chemistry .2. Calibration and Regression Methods - A Review. Analyst 116: pp 3-14. Mitchison D.A. and Fourie P.B. (2010) The Near Future: Improving the Activity of Rifamycins and Pyrazinamide. Tuberculosis 90: pp 177-181. Moghimi S.M. and Hunter A.C. (2001) Recognition by Macrophages and Liver Cells of Opsonized Phospholipid Vesicles and Phospholipid Headgroups. Pharmaceutical Research 18: pp 1-8. Nuermberger E, and Grosset J. (2004) Pharmacokinetic and Pharmacodynamic Issues in the Treatment of Mycobacterial Infections. European Journal of Clinical Microbiology & Infectious Diseases 23: pp 243-255. Pedrosa J., Florido M., Kunze Z.M., Castro A.G., Portaels F., Mcfadden J., Silva M.T. and Appelberg R. (1994) Characterization of the Virulence of Mycobacterium-Avium Complex (Mac) Isolates in Mice. Clinical and Experimental Immunology 98: pp 210-216. Pinto-Alphandary H., Andremont A. and Couvreur P. (2000) Targeted Delivery of Antibiotics Using Liposomes and Nanoparticles: Research and Applications. International Journal of Antimicrobial Agents 13: pp 155-168. Pires, D. Role of mycobacteria porins and efflux pumps on the intracellular survival within macrophages. 2011. Faculdade de Ciências da Universidade de Lisboa . Rautio J., Kumpulainen H., Heimbach T., Oliyai R., Oh D., Jarvinen T. and Savolainen J. (2008) Prodrugs: Design and Clinical Applications. Nature Reviews Drug Discovery 7: pp 255-270. Raynaud C., Laneelle M.A., Senaratne R.H., Draper P., Laneelle G. and Daffe M. (1999) Mechanisms of Pyrazinamide Resistance in Mycobacteria: Importance of Lack of Uptake in Addition to Lack of Pyrazinamidase Activity. Microbiology-Uk 145: pp 1359-1367. Reed C., von Reyn C.F., Chamblee S., Ellerbrock T.V., Johnson J.W., Marsh B.J., Johnson L.S., Trenschel R.J. and Horsburgh C.R. (2006) Environmental Risk Factors for Infection With Mycobacterium Avium Complex. American Journal of Epidemiology 164: pp 32-40. Ribeiro M.M.B., Melo M.N., Serrano I.D., Santos N.C. and Castanho M.A.R.B. (2010) Drug-Lipid Interaction Evaluation: Why a 19th Century Solution? Trends in Pharmacological Sciences 31: pp 449-454. Rouser G., Fleische S. and Yamamoto A. (1970) 2 Dimensional Thin Layer Chromatographic Separation of Polar Lipids and Determination of Phospholipids by Phosphorus Analysis of Spots. Lipids 5: pp 494-&. Sarkar S., Mandal S., Sinha J., Mukhopadhyay S., Das N. and Basu M.K. (2002) Quercetin: Critical Evaluation As an Antileishmanial Agent In Vivo in Hamsters Using Different Vesicular Delivery Modes. Journal of Drug Targeting 10 (8): pp 573-578.

50

Silva M.T., Appelberg R., Silva M.N.T. and Macedo P.M. (1987) Invivo Killing and Degradation of Mycobacterium-Aurum Within Mouse Peritoneal-Macrophages. Infection and Immunity 55: pp 2006-2016. Simões, M. F. Estudo da estabilidade e actividade de pró-fármacos derivados da pirazinamida sob a forma livre e encapsulada em lipossomas. 2005. Universidade Lusófona de Humanidades e Tecnologia. Simões M.F., Valente E., Gomez M.J.R., Anes E. and Constantino L. (2009) Lipophilic Pyrazinoic Acid Amide and Ester Prodrugs Stability, Activation and Activity Against M. Tuberculosis. European Journal of Pharmaceutical Sciences 37: pp 257-263. Taylor K.M.G. and Morris R.M. (1995) Thermal-Analysis of Phase-Transition Behavior in Liposomes. Thermochimica Acta 248: pp 289-301. Torchilin V.P. (2005) Recent Advances With Liposomes As Pharmaceutical Carriers. Nature Reviews Drug Discovery 4: pp 145-160. Valente E., Simoes M.F., Testa B. and Constantino L. (2011) Development of a Method to Investigate the Hydrolysis of Xenobiotic Esters by a Mycobacterium Smegmatis Homogenate. Journal of Microbiological Methods 85: pp 98-102. Wade M.M. and Zhang Y. (2004) Anaerobic Incubation Conditions Enhance Pyrazinamide Activity Against Mycobacterium Tuberculosis. Journal of Medical Microbiology 53: pp 769-773. Wolinsky E. (1979) Non-Tuberculous Mycobacteria and Associated Diseases. American Review of Respiratory Disease 119: pp 107-159. Yamamoto S., Toida I., Watanabe N. and Ura T. (1995) In-Vitro Antimycobacterial Activities of Pyrazinamide Analogs. Antimicrobial Agents and Chemotherapy 39: pp 2088-2091. Yardley V. and Croft S.L. (2000) A Comparison of the Activities of Three Amphotericin B Lipid Formulations Against Experimental Visceral and Cutaneous Leishmaniasis. International Journal of Antimicrobial Agents 13: pp 243-248. Zar, J.H. (2009) Biostatistical Analysis. 5

Th Ed., Prentice Hall, London

Zhang Y. and Mitchison D. (2003) The Curious Characteristics of Pyrazinamide: a Review. International Journal of Tuberculosis and Lung Disease 7: pp 6-21. Zhang Y., Wade M.M., Scorpio A., Zhang H. and Sun Z.H. (2003) Mode of Action of Pyrazinamide: Disruption of Mycobacterium Tuberculosis Membrane Transport and Energetics by Pyrazinoic Acid. Journal of Antimicrobial Chemotherapy 52: pp 790-795.