• Non ci sono risultati.

On the possibility of low cost, adherent therapeutic drug monitoring in oncology

N/A
N/A
Protected

Academic year: 2021

Condividi "On the possibility of low cost, adherent therapeutic drug monitoring in oncology"

Copied!
6
0
0

Testo completo

(1)

On the possibility of low cost, adherent Therapeutic Drug Monitoring

in Oncology

Silvia Dalla Marta

a

, Stefano Fornasaro

a

, Aleksandra Jaworska

a

, Giuseppe Toffoli

b

,

Alois Bonifacio

a

° and Valter Sergo

a

°

a

Raman Spectroscopy group, Dept. of Architecture and Engineering, University of Trieste, I-34127

Trieste, ITALY;

b

Experimental and Clinical Pharmacology Division, Department of Translational

Research, IRCCS-National Cancer Institute, Aviano, ITALY; °Co-last authors.

ABSTRACT

A frequent quantification of drugs concentrations in plasma of patients subject to chemotherapy is seldom performed, mostly because the standard methods (Gas or Liquid Chromatography coupled with Mass Spectroscopy) are expensive and time consuming.

In this paper we report the approach pursued in one of the research units of the EU project RAMAN4CLINICS to tackle the problem of a low cost, time adherent quantification of drugs used for oncological patients using a Surface Enhanced Raman Scattering (SERS) spectroscopy.

More specifically, the issues concerning the repeatability of the nanostructured substrates will be presented and some promising results to increase the selectivity of the measures toward specific drugs will be discussed, with examples concerning one cytotoxic agent, Irinotecan and one kinase inhibitor, Sunitinib.

Keywords: SERS, Therapeutic drug monitoring, paper substrate

1. INTRODUCTION

Chemotherapeutic drugs are expensive, have rather heavy side effects on patients, and are usually effective in a very narrow therapeutic window of plasmatic concentrations. All these factors point to the opportunity to optimize their use, adjusting in real time the dosage of the drugs administered to the patients in what is called Therapeutic Drug Monitoring (TDM). Unfortunately, the golden standard for the quantification of drugs in the course of chemotherapeutic treatments is Mass Spectroscopy coupled with High Performance Liquid Chromatography (HPLC-MS), which is both time consuming and expensive. Consequently, TDM is seldom performed on oncological patients and the administration of the drugs is grossly adjusted on the base of the side effects as reported by the patients or as inferred from the routine blood analysis. Simple, rapid and low-cost methods for chemotherapeutic drugs quantification are thus needed to effectively implement TDM. In this contribution we outline an approach that we are pursuing to reach this goal by using a combination of Raman spectroscopy, nanotechnology and chemometrics. Raman spectroscopy in particular has long attracted the interest of the medical community as an analytical tool and it is at the heart of a European initiative called RAMAN4CLINICS, aimed at introducing Raman spectroscopy in clinical practice. One of the activities of RAMAN4CLINICS is specifically devoted to TDM, and the approach used constitutes the backbone of this contribution. When molecules are absorbed on nanostructured metal surfaces, their Raman spectral signature is highly enhanced: this effect is known as Surface Enhanced Raman Scattering (SERS), where the enhancement is up to billions of time (with respect to the spectrum of the same analyte with the same concentration but without adsorption on the nanostructured metal surface). The strong SERS enhancement can be exploited to obtain information from chemotherapeutic drugs in concentration of interest for the oncological patients, typically from nanomolar to micromolar. SERS approaches for drug quantification have been previously tried, also using a separation step different than HPCL (e.g. TLC) before SERS analysis [1]. Once the SERS spectra are obtained, several chemometric techniques can be applied to obtain calibration curves/models capable of establishing appropriate relationships between instrumental responses and the concentration of the drugs of interest in blood/plasma/serum of the patients, which is the ultimate goal.

Biophotonics: Photonic Solutions for Better Health Care V, edited by Jürgen Popp, Valery V. Tuchin, Dennis L. Matthews, Francesco Saverio Pavone, Proc. of SPIE Vol. 9887, 98870G

© 2016 SPIE · CCC code: 0277-786X/16/$18 · doi: 10.1117/12.2225232

Proc. of SPIE Vol. 9887 98870G-1

(2)

A flowchart o boxes of the f

Figure 1 F chem

2.1 SERS Su The solid SER (Ag) NPs on Lee Meisel m capacity of 1 concentration aggregation. such time, al plastic syring air at room te feature an int of the propose flowchart, refe Flowchart of the mometric analys ubstrate prep RS substrates common filte method, respe 0 mL) contai n of 20 mM.

The vials con ll the NPs we ge, paying atte emperature an tra- and

inter-ed approach in ferring the inte

e approach prop sis. paration used in this p er paper with ectively [4]. ining 3 mL o The presence ntaining the fil

ere deposited ention to not t nd then stocke sample repeat

2. ME

n presented in erested reader posed to achiev paper were ob a 2 µm pore A piece of 1 f Au or Ag c of the citrate lter paper wer on the vial b touch or move ed in Milli-Q tability of 15%

ETHODOL

n Figure 1. In to more speci ve a TDM of ch tained using t size. Au and 1 cm2 was pl colloidal solut e leaded to a re then stocke bottom, cover e the paper su water to avoi % for Au NPs

LOGY

this section w ific literature hemotherapeutic the dip-coating Ag NPs were aced on the b tion, to which color change ed in the dark ring the paper ubstrate. The d any loss of and 20% for we will presen for details. c drugs through g method [2], e synthetized bottom of a c h sodium citra of the colloi at room temp r. The supern substrates we NP plasmoni Ag NPs. nt some data f h SERS spectros loading gold with the Turk cylindrical gl ate was added idal solution, perature for on natant was ext

re then dried ic activity. Th

for each of the

scopy and

(Au) or silver kevich [3] and lass vial (tota d up to a fina indicating NP ne week; after tracted with a in the vials in hose substrates e r d al al P r a n s

Proc. of SPIE Vol. 9887 98870G-2

(3)

Figure 2 S 2.2 Adsorpt Preliminary S surrogate ma similar to tho during such t acquiring the minus one int drug concent necessary, th biofluids such of the matrix. Figure 3 S and S

Solid SERS sub

tion of drugs SERS measur atrix frequently

ose of the biof time the mole e SERS spectr tensity standa tration detecte erefore, to inc h as serum an . Solid Au SERS SERS spectra o bstrate developm on substrate rements of an y used to sim fluids. The su ecules adsorb ra of the drug ard deviation, a ed it is still to crease the sub nd/or plasma, i substrate imag of Irinotecan in ment process. nticancer drug mulate plasma ubstrates were b on the NPs gs. Figure 3 re acquired on A oo high comp bstrates sensit in which drug ge, cartoon of m a concentration gs were perfor and serum, b e then incubat surface. The eports the ave Au solid SERS pared with the ivity especial g molecules m molecular adsorp n of 270 µM, ac rmed in Phos because its pH ted with the a

substrates we erage SERS sp S substrates pr e typical plasm ly if the antic must compete w ption of NPs su cquired on Au s sphate-Buffere H (7.4) and io anticancer drug ere allowed to pectrum of Ir repared as des matic values i cancer drug m with virtually urfaces, Irinotec solid SERS subs

ed Saline (PB onic strength (

g solution for o dry and fin rinotecan 270 scribed in 2.1. in oncologica must be detecte

all of the othe

can molecular st strate. BS) solution, a (150 mM) are r few minutes nally analyzed µM, plus and However, the al patient. It is ed in complex er components tructure a e s; d, d e s x s

Proc. of SPIE Vol. 9887 98870G-3

(4)

2.3 Coating One of the im substrates to functionalized supramolecul symmetric m [5]. We exp Irinotecan (fi bare NPs in a with (CB[7]) concentration Figure 4 S adsor Irino Irino 2.4 Chemom To monitor th which is poss calibration, s intensities (or value to conc the instrumen of SERS sub mportant chal match the th d the NPs s lar host-guest macrocyclic. Du loited these r igure 4). In pa a concentratio ), in a concen n of two order Solid Au SERS rption process o otecan in a conc otecan in a con metric analysi he plasmatic c sible to extrap spectra of sam r band areas) centration. Alt ntal response bstrates with llenges for ap herapeutic ran surfaces, hen interactions. ue to this barr recognition m articular, we on of 270 µM ntration of 2. s of magnitud substrate imag on NPs surface centration of 27 ncentration of 2 is for obtaini concentration polate the info mples with kn are plotted a though univar to depend on “recognition” pplication in c ge of the ant nce the solid Cucurbit[n]ur rel-like shape, molecules to report the SE , and ii) the S .7 µM. In th de lower. ge, cartoon of in . Irinotecan and 0 µM, acquired 2.7 µM, acquire ing calibratio of anticancer ormation abou nown concentr s a function o riate calibratio nly on the co ” elements clinical setting ticancer drugs d SERS subs rils (CB[n]), a , those host st increase the ERS spectra o SERS spectra e presence of ntermolecular in d curcubit[7]uri d on bare Au so ed on function on models r drugs in biof ut the concent rations of ana of concentrati on is the most oncentration o gs is the impr s (typically do strates, with are a family o tructures are a sensitivity o f the anticanc of the same m f (CB[7]), th nteraction of Iri il molecular stru olid SERS subs nalized Au solid fluids it is nec tration of the alytes are me on and a regr intuitive and of the analyte rovement of t own to 0.01 m recognition of supramolec able to entrap f the substra cer drug acqu molecule acqu e anticancer

inotecan and (C uctures. On the strate (in blue) d SERS substra cessary to per anticancer dru easured. In a u ression is perf simple appro e of interest. the sensitivity mM). For thi molecules, e cular hosts, wi guest molecu ates for the c uired in PBS s uired on funct drug can be

CB[7]) and the r left: SERS spe ) and SERS spe ate (in grey).

rform a calibr ug from SERS

univariate app formed relatin

ach, its applic Interferences

y of our SERS s purpose, we exploiting the ith a rigid and ules in solution cytotoxic drug solutions i) on tionalized NPs detected in a related ectra of ectra of ration, through S spectra. In a proach, single ng a measured cation requires s from severa S e e d n g n s a h a e d s al

Proc. of SPIE Vol. 9887 98870G-4

(5)

1500- 1000- 500-Univar alibration O

A

O 0 o O o 0 00 0 o 8 I! 0 O dv 0 m8 0 00 0 0 0 0 8 o O 0 O e 50 100 MTX concentration (pM) O o 150 400 560 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 Raman Shift (cm -1) Multiv to calibration (PLSR)

serum/plasma components may contribute to the measured intensity at a specific Raman shift, making often impossible to differentiate an analyte-specific signal from an interfering one when looking only at one point of a data spectrum. An alternative is a multivariate calibration, using multiple responses simultaneously (e.g., the response at a range of Raman shifts, or over the entire range collected, to calculate concentrations), which may overcome most of the limitations of univariate calibration. In figure 5 we report promising preliminary results of Methotrexate (MTX) quantification in diluted human serum with SERS substrate and chemometric analysis: multivariate calibration models built using partial least-squares regression (PLSR) were compared with univariate calibration. The PLS model outperformed the univariate model.

Figure 5 Diagnostic plots from univariate and multivariate calibration for SERS spectra of MTX collected in diluted human serum. A: (o) calibration data for the univariate calibration, confidence bands (broken blue line) and prediction bands (broken red line). B: results from the 100 repetitions in Repeated Double Cross Validation (RDCV) for a Partial Least Square Regression (PLSR) are reported in orange; the means of 91 predictions are reported in yellow. SERS spectrum of MTX 10 μM and the chemical structure of MTX are reported for reference. All spectra were collected on

functionalized Au solid SERS substrate using an excitation at 785 nm.

Proc. of SPIE Vol. 9887 98870G-5

(6)

3. CONCLUSIONS

In this work, we present solid SERS substrates of Au and Ag NPs, developed with a simple dip-coating method, able to quantify anticancer drugs in a model PBS solution and in a complex matrix, such as diluted human serum, as well if the SERS technique is combined with multivariate calibration. However, the substrates sensitivity its still too low to detect some anticancer agent, such as Irinotecan, in biofluids in therapeutic plasmatic values and moreover, the observed prediction error obtained with multivariate calibration it is still too high if compared with the value provided by established reference methods. Thus, an increase in selectivity and sensitivity of the SERS substrates is necessary. For this reason, we present preliminary results of substrates functionalization using the host-guest supramolecular approach, which increase the substrates sensitivity, at least in a PBS solution, of Irinotecan concentration detection of 2 orders of magnitude. Moreover, we show the advantages of multivariate regression methods with respect to univariate calibration, in the case of MTX.

Overall, this contribution reports the logical procedure used in one of the approaches of the Working Group 1 (Therapeutic monitoring of antitumoral drugs and antibiotic in body fluids) of the EU COST (European Cooperation in Science and Technology) action RAMAN4CLINICS.

AKNOWLEDGMENTS

SF acknowledges support from IRCCS Burlo Garofolo. AB and, AJ and VS acknowledge support from AIRC (project code 12214), and EU COST action BM1401 (Raman4clinics).

REFERENCES

[1] A. Vicario, V. Sergo, G. Toffoli et al., “Surface-enhanced Raman spectroscopy of the anti-cancer drug irinotecan in presence of human serum albumin,” Colloids and surfaces. B, Biointerfaces, 127C, 41-46 (2015). [2] L. Polavarapu, and L. M. Liz-Marzan, “Towards low-cost flexible substrates for nanoplasmonic sensing,” Phys

Chem Chem Phys, 15(15), 5288-300 (2013).

[3] J. Turkevich, P. C. Stevenson, and J. Hillier, “A study of the nucleation and growth processes in the synthesis of colloidal gold,” Discuss. Faraday Soc., 11(0), 55-75 (1951).

[4] P. C. Lee, and D. Meisel, “Adsorption and surface-enhanced Raman of dyes on silver and gold sols,” The Journal of Physical Chemistry, 86(17), 3391-3395 (1982).

[5] S. Mahajan, T. C. Lee, F. Biedermann et al., “Raman and SERS spectroscopy of cucurbit[n]urils,” Phys Chem Chem Phys, 12(35), 10429-33 (2010).

Proc. of SPIE Vol. 9887 98870G-6

Riferimenti

Documenti correlati

In this study, we demonstrated for the first time that information extracted from communicative gestures influences the concurrent processing of biological motion by

For the particular case considered here (CRs outside the cloud have a spectrum equal to that observed by Voyager), it is clear that the differential ionization rate peaks at about

Enhanced Raman Scattering of Rhodamine 6G Films on Two-Dimensional Transition Metal Dichalcogenides Correlated to Photoinduced Charge Transfer. Raman Enhancement Effect

tecniche di imaging, descritte brevemente nel seguito, richiedono molte volte degli agenti di contrasto e il presente lavoro di tesi è rivolto alla sintesi di un sistema

Transthoracic echocardiogra- phy (TTE) showed moderate aortic root di- latation (at sinus of Valsalva: 44 mm) com- pared to normal references for age and body surface area and

In this randomized study involving patients with newly diagnosed multiple myeloma, the stan- dard high-dose melphalan consolidation therapy followed by stem-cell transplantation,

In the actual cutting process, the material removed and the diamond bead wear depend on the following main process parameters: pulling force and wire speed.. Both are controllable

Furthermore, we analysed p65BTK expression in a cohort of 83 stage II colon carcinoma patients and found that in 68.7% of peritumoural/tumoural sample pairs, p65BTK was more